WO2018231973A1 - Inhibitors of ezh2 and methods of use thereof - Google Patents

Inhibitors of ezh2 and methods of use thereof Download PDF

Info

Publication number
WO2018231973A1
WO2018231973A1 PCT/US2018/037321 US2018037321W WO2018231973A1 WO 2018231973 A1 WO2018231973 A1 WO 2018231973A1 US 2018037321 W US2018037321 W US 2018037321W WO 2018231973 A1 WO2018231973 A1 WO 2018231973A1
Authority
WO
WIPO (PCT)
Prior art keywords
mutation
ezh2
subject
cancer
function
Prior art date
Application number
PCT/US2018/037321
Other languages
French (fr)
Inventor
Stephen Blakemore
Scott Richard Daigle
Original Assignee
Epizyme, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Epizyme, Inc. filed Critical Epizyme, Inc.
Priority to US16/622,391 priority Critical patent/US20230201212A1/en
Publication of WO2018231973A1 publication Critical patent/WO2018231973A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the disclosure provides a method of treating cancer comprising administering a therapeutically effective amount of an inhibitor of Enhancer to Zeste Homolog 2 (EZH2) to a subject in need thereof, wherein the cancer is characterized by at least one mutation in one or more sequences encoding a gene or gene product listed in Tables 1 -5, and/or Figures 1- 3.
  • the cancer is characterized by at least one mutation in one or more sequences encoding: EZH2, MYD88, STAT6A, MYC, TP53, and/or HIST1H1E.
  • the cancer is characterized by a genotype that satisfies Genetic Profile 1 of Figure 3.
  • the cancer is characterized by a genotype that satisfies Genetic Profile 2 of Table 5.
  • the cancer comprises a MYD88 gain-of-function mutation.
  • the cancer does not comprise an EZH2 gain-of-function mutation.
  • the at least one mutation decreases the function of a protein encoded by the mutated sequence as compared to the function of the protein encoded by the wild-type sequence.
  • the at least one mutation is a loss-of-function mutation.
  • the method further comprises detecting the at least one mutation in the subject.
  • the detecting comprises subjecting a sample obtained from the subject to a sequence analysis assay.
  • the analysis assay is a cobas® assay.
  • the detecting comprises obtaining a sample from the subject.
  • the sample is a tumor sample or a sample that comprises a cancer cell. In some embodiments, the sample is a sample comprising ctDNA. In some embodiments, the inhibitor of
  • the inhibitor of EZH2 is administered orally. In some embodiments, the inhibitor of EZH2 is administered orally.
  • the inhibitor of EZH2 is formulated as a tablet. In some embodiments, the therapeutically effective amount of the inhibitor of EZH2 is between 100 mg and 3200 mg per day. In some embodiments, the therapeutically effective amount of the inhibitor of EZH2 is 100 mg, 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, 1400 mg, 1600 mg or 3200 mg per day. In some embodiments, the therapeutically effective amount is 1600 mg per day. In some embodiments, the cancer expresses a wild type EZH2 protein and does not express a mutant
  • the cancer expresses a mutant EZH2 protein.
  • the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1.
  • the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1.
  • EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 682 of SEQ ID NO: 1.
  • the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 692 of SEQ ID NO: 1.
  • the at least one mutation comprises a MYD88 and/or STAT6A mutation, preferably wherein the MYD88 mutation is a gain-of-function mutation, preferably a
  • the cancer does not have a MYC and/or a
  • the cancer (a) has a MYD88 gain-of-function or a
  • the cancer (a) has a MYD88 L273 mutation or a STAT6A loss-of-function mutation, (b) has neither a MYC nor a HIST1H1E mutation, and (c) does not have an EZH2 gain-of-function mutation.
  • the subject is a human subject.
  • the subject has cancer.
  • the cancer is B-cell lymphoma.
  • the B-cell lymphoma is an activated B-cell (ABC) type.
  • the B-cell lymphoma is a germinal B-cell (GBC) type.
  • the cancer is follicular lymphoma.
  • Some aspects of this disclosure provide methods comprising selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of at least one mutation associated with a positive response to such treatment in the subject and/or based on the absence of at least one mutation associated with no response or with a negative response to such treatment in the subject.
  • the at least one mutation associated with a positive response comprise (a) an EZH2 gain-of-function mutation or a MYD88 gain-of-function mutation; and (b) a STAT6 loss-of-function mutation.
  • the at least one mutation associated with no response or with a negative response comprise (a) a MYC mutation; (b) a TP53 mutation; and/or (c) a HIST1H1E mutation.
  • the method comprises detecting the at least one mutation associated with a positive response and/or the at least one mutation associated with no response or a negative response in a sample obtained from the subject.
  • the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having either an EZH2 or a MYD88 gain-of-function mutation, or a STAT6A loss-of-function mutation; and (b) having neither a MYC mutation, nor a TP53 mutation, nor a HIST1H1E mutation.
  • the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having a MYD88 L273 mutation or a STAT6A mutation, and (b) having neither a MYC mutation, nor a TP53 mutation, nor a HIST1H1E mutation.
  • the at least one mutation consists of a single mutation. In some embodiments, the at least one mutation comprises 2 mutations or more. In some embodiments, the at least one mutation comprises 3 mutations or more. In some embodiments, the at least one mutation comprises 4 mutations or more.
  • Some aspects of this disclosure provide methods comprising selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of a mutation profile in the subject that comprises one or more mutations described for a patient exhibiting a complete or partial response or stable disease in any of Tables 1-5 or Figures 1-3, or that comprises, encompasses, or matches the complete mutation profile of such a patient, or that comprises, encompasses, or matches Genetic Profile 1 or Genetic Profile 2 of Table 5. .
  • Exemplary sequences associated with the genes and proteins provided herein include, without limitation: MYD88 (e.g., GenBank Accession No. NM OOl 172567.1, NM 002468.4, NM_001172568.1, NM_001172569.1, and NM_001172566.1), STAT6A (e.g., GenBank Accession No. NM OO 1178078.1, NM 003153.4, NM_001178079.1, NM OO 1178080.1, or NM_001178081.1), SOCS1 (e.g., GenBank Accession No. NM_003745.1), MYC (e.g., GenBank Accession No. NM 002467.4), fflSTlHlE (e.g., GenBank Accession No.
  • NM_005321.2 ABLl (e.g., GenBank Accession No. NM_005157), ACVR1 (e.g., GenBank Accession No. NM_001105.4), AKTl (e.g., GenBank Accession No. NM_001014431.1), AKT2 (e.g., GenBank Accession No. NM 001243027.2), ALK (e.g., GenBank Accession No.
  • APC e.g., GenBank Accession No. NM_000038.5
  • AR e.g., GenBank Accession No. NM 000044.3
  • ARID1A e.g., GenBank Accession No. NM 006015.4
  • ARID IB e.g., GenBank Accession No. NM 020732.3
  • ASXL1 e.g., GenBank Accession No. NM_015338.5
  • ATM e.g., GenBank Accession No. NM_000051.3
  • ATRX e.g., GenBank Accession No. NM_000489.4
  • AURKA e.g., GenBank Accession No. NM_003600.3
  • ⁇ 2 e.g., GenBank Accession No. NM 004655.3
  • BAPl e.g., GenBank Accession No.
  • NM_004656.3 BCL2 (e.g., GenBank Accession No. NM_000633.2), BCR (e.g., GenBank Accession No. X02596.1), BLM (e.g., GenBank Accession No. NM_000057.3), BMPR1A (e.g., GenBank Accession No. NM 004329.2), BRAF (e.g., GenBank Accession No. NM 004333.4), BRCA1 (e.g., GenBank Accession No. NM_007294.3), BRCA2 (e.g., GenBank Accession No. NM_000059.3), BRIPl (e.g., GenBank Accession No.
  • NM_032043.21 BTK (e.g., GenBank Accession No. NM_001287344.1), BUB IB (e.g., GenBank Accession No. NM_001211.5), CALR (e.g., GenBank Accession No. NM 004343.3), CBL (e.g., GenBank Accession No. NM_005188.3), CCND1 (e.g., GenBank Accession No. NM_053056.2), CCNE1 (e.g., GenBank Accession No. NM_001322262.1), CDC73 (e.g., GenBank Accession No. NM_024529.4), CDH1 (Accession No. NM_001317186.1), CDK4 (e.g., GenBank Accession No.
  • CDK6 e.g., GenBank Accession No. NM OOl 145306.1
  • CDKN1B e.g., GenBank Accession No. NM 004064.4
  • CDKN2A e.g., GenBank Accession No.
  • CDKN2B e.g., GenBank Accession No. NM 078487.2
  • CDKN2C e.g., GenBank Accession No. NM 078626.2
  • CEBPA e.g., GenBank Accession No. NM_001285829.1
  • CHEK2 e.g., GenBank Accession No. NM_145862.2
  • CIC e.g., GenBank Accession No. NM_015125.4
  • CREBBP e.g., GenBank Accession No. NM_001079846.1
  • CSF1R e.g., GenBank Accession No.
  • NM 001288705.2 CTNNB1 (e.g., GenBank Accession No. NM_001098209.1), CYLD (e.g., GenBank Accession No. NM 001042355.1), DAXX (Accession No. NM 001141969.1), DDB2 (e.g., GenBank Accession No. NM 001300734.1), DDR2 (e.g., GenBank Accession No. NM 001014796.1), DICERl (e.g., GenBank Accession No. NM_001291628.1), DNMT3A (e.g., GenBank Accession No. NM 001320893.1), EGFR (e.g., GenBank Accession No. NM 001346900.1), EP300 (e.g., GenBank Accession No.
  • ERBB2 e.g., GenBank Accession No. NM_001289936.1
  • ERBB3 e.g., GenBank Accession No. NM OO 1982.3
  • ERBB4 e.g., GenBank Accession No.
  • ERCC1 e.g., GenBank Accession No. NM 001166049.1
  • ERCC2 e.g., GenBank Accession No. NM 001130867.1
  • ERCC3 e.g., GenBank Accession No.
  • ERCC4 Accession No. NM_005236.2
  • ERCC5 e.g., GenBank Accession No. NM_000123.3
  • ESR1 e.g., GenBank Accession No. NM_001291241.1
  • ETV1 e.g., GenBank Accession No. NM_001163147.1
  • ETV5 e.g., GenBank Accession No. NM_004454.2
  • EWSR1 e.g., GenBank Accession No. NM 001163287.1
  • EXT1 e.g., GenBank Accession No.
  • NM_000127.2 EXT2 (Accession No. NM_001178083.1), FANCA (e.g., GenBank Accession No. NM_001286167.1), FANCB (Accession No. NM 001324162.1), FANCC (e.g., GenBank Accession No. NM_001243744.1), FANCD2 (e.g., GenBank Accession No. NM_001319984.1), FANCE (e.g., GenBank Accession No. NM_021922.2), FANCF (e.g., GenBank Accession No NM 022725.3.), FANCG (e.g., GenBank Accession No. NM_004629.1), FANCI (e.g.,
  • GenBank Accession No. NM_018193.2 FANCL (Accession No. NM_001114636.1), FANCM (e.g., GenBank Accession No. NM 001308133.1), FBXW7 (e.g., GenBank Accession No. NM_018315.4), FGFR1 (Accession No.) NM_001174065.1, FGFR2 (e.g., GenBank Accession No. NM_000141.4), FGFR3 (e.g., GenBank Accession No. NM_001163213.1), FGFR4 (e.g., GenBank Accession No. NM 213647.2), FH (e.g., GenBank Accession No.
  • NM 000143.3 FLCN (e.g., GenBank Accession No. NM 144606.5), FLT3 (e.g., GenBank Accession No. NM_004119.2), FLT4 (e.g., GenBank Accession No. NM_002020.4), FOXL2 (e.g., GenBank Accession No. NM_023067.3), GATA1 (e.g., GenBank No. NM_002049.3), GATA2 (e.g., GenBank Accession No. NM OOl 145662.1), GNA11 (e.g., GenBank Accession No.
  • NM_002067.4 e.g., GenBank Accession No. NM_002072.4
  • GNAS e.g., GenBank Accession No. NM_080425.3
  • GPC3 e.g., GenBank Accession No. NM_001164619.1
  • H3F3A e.g., GenBank Accession No. NM 002107.4
  • H3F3B e.g., GenBank Accession No. NM_005324.4
  • HNF1A e.g., GenBank Accession No. NM_000545.6
  • HRAS e.g., GenBank Accession No. NM_001130442.2
  • IDH1 e.g., GenBank Accession No.
  • IDH2 e.g., GenBankAccessionNo. NM_001290114.1
  • IGF1R e.g., GenBank Accession No. NM_001291858.1
  • IGF2R e.g., GenBank Accession No. NM 000876.3
  • KZFl e.g., GenBank AccessionNo. NM 001291847.1
  • JAK1 e.g., GenBank Accession No.
  • JAK2 e.g., GenBank AccessionNo. NM_001322195.1
  • JAK3 e.g., GenBank Accession No. NM 000215.3
  • KDR e.g., GenBank Accession No. NM 002253.2
  • KIT e.g., GenBank Accession No. NM 001093772.1
  • KRAS e.g., GenBank Accession No. NM_033360.3
  • MAML1 e.g., GenBank Accession No. NM_014757.4
  • MAP2K1 e.g., GenBank Accession No. NM 002755.3
  • MAP2K4 e.g., GenBank Accession No.
  • MDM2 e.g., GenBank Accession No. NM_001145337.2
  • MDM4 e.g., GenBank Accession No. NM 001278519.1
  • MED12 e.g., GenBank Accession No.
  • NM_005120.2 e.g., GenBank Accession No. NM_130804.2
  • MET e.g., GenBank Accession No NM 000245.3
  • MLH1 e.g., GenBank Accession No. NM 000249.3
  • MLL e.g., GenBank AccessionNo. AF232001.1
  • MPL e.g., GenBank AccessionNo.
  • NM_005373.2 e.g., GenBank Accession No. NM_000251.2
  • MSH6 e.g., GenBank Accession No. NM_000179.2
  • MTOR e.g., Accession No. NM_004958.3
  • MUTYH e.g., GenBank Accession No. NM 001048171.1
  • MYC e.g., GenBank Accession No.
  • MYCL1 e.g., GenBank Accession No NM 001033081.2
  • MYCN e.g., GenBank Accession No. NM 001293231.1
  • NBN e.g., GenBank Accession No.
  • NCOA3 e.g., GenBank Accession No. NM OOl 174087.1
  • NFl e.g., GenBank Accession No. NM OO 1042492.2
  • NF2 e.g., GenBank Accession No.
  • NKX2-l e.g., GenBank Accession No. NM_001079668.2
  • NOTCH1 e.g., GenBank Accession No. NM 017617.4
  • NOTCH2 e.g., GenBank Accession No
  • NM_001200001.1 e.g., GenBank Accession No. NM_000435.2
  • NOTCH4 e.g., NR_134950.1
  • NPM1 e.g., GenBank Accession No. NM_002520.6
  • NRAS Accession No. NM_002524.4
  • NTRKl e.g., GenBank AccessionNo. NM_001007792.1
  • PALB2 e.g., GenBank Accession No. NM 024675.3
  • PAX5 e.g., GenBank Accession No. NM 001280552.1
  • PBRMl e.g., GenBank AccessionNo. NM_181042.4
  • PDGFRA e.g., GenBank Accession No. NM 006206.4
  • PHOX2B e.g., GenBank Accession No.
  • PIK3CA e.g., GenBank Accession No. NM_006218.3
  • PIK3R1 e.g., PIK3R1
  • PMS1 e.g., GenBank Accession No. NM_001321051.1
  • PMS2 e.g., GenBank Accession No. NM 000535.6
  • POLD1 e.g., GenBank Accession No.
  • NM 001308632.1 POLE (e.g., GenBank Accession No. NM_006231.3), POLH (e.g., GenBank Accession No. NM_001291970.1), POT1 (e.g., GenBank Accession No. NM_001042594.1), PRKAR1A (e.g., GenBank Accession No. NM 001278433.1), PRSS1 (e.g., GenBank Accession No. NM_002769.4), PTCH1 (e.g., GenBank Accession No. NM_000264.3), PTEN (e.g., GenBank Accession No. NM 000314.6), PTPN11 (e.g., GenBank Accession No.
  • NM_001330437.1 RAD51C (e.g., GenBank Accession No. NR_103873.1), RAF1 (e.g., GenBank Accession No. NM 002880.3), RBI (e.g., GenBank Accession No. NM 000321.2), RECQL4 (e.g., GenBank Accession No. NM 004260.3), RET (e.g., GenBank Accession No.), RNF43(e.g., GenBank Accession No. NM_017763.5), ROS1 (e.g., GenBank Accession No. NM 002944.2), RUNX1 (e.g., GenBank Accession No. NM_001122607.1), SBDS (e.g., GenBank Accession No. NM 016038.2), SDHAF2 (e.g., GenBank Accession No.
  • SDHB e.g., GenBank Accession No.
  • SDHC e.g., GenBank Accession No.
  • SDHD e.g., GenBank Accession No. NM 001276503.1
  • SF3B1 e.g., GenBank Accession No. NM 001308824.1
  • SMAD2 e.g., GenBank Accession No. NM_001135937.2
  • SMAD3 e.g., GenBank Accession No. NM 001145104.1
  • SMAD4 e.g., GenBank Accession No.
  • SMARCB1 e.g., GenBank Accession No. NM_001007468.2
  • SMO e.g., GenBank Accession No. NM 005631.4
  • SRC e.g., GenBank Accession No. NM 005417.4
  • STAG2 e.g., GenBank Accession No. NM_001282418.1
  • STKll e.g., GenBank Accession No. NM_000455.4
  • SUFU e.g., GenBank Accession No. NM_001178133.1
  • TERT e.g., GenBank Accession No. NM_001193376.1
  • TET2 e.g., GenBank Accession No.
  • TGFBR2 e.g., GenBank Accession No. NM_001024847.2
  • TNFAIP3 e.g., GenBank Accession No. NM OO 1270508.1
  • TOPI e.g., GenBank Accession No.
  • TP53 e.g., GenBank Accession No. NM_000546.5
  • TSC1 e.g., GenBank Accession No. NM_001162427.1
  • TSC2 e.g., GenBank Accession No. NM_001318832.1
  • TSHR e.g., GenBank Accession No. NM_000369.2
  • VHL e.g., GenBank Accession No. NM_000551.3
  • WAS e.g., GenBank Accession No. NM 000377.2
  • WRN e.g., GenBank Accession No. NM_000553.4
  • WT1 e.g., GenBank Accession No.
  • NM_000378.4 e.g., GenBank Accession No. NM 000380.3
  • XPC e.g., GenBank Accession No. NM 004628.4
  • XRCC1 e.g., GenBank Accession No. NM 006297.2
  • the at least one mutation decreases the function of a protein encoded by the mutated sequence as compared to the function of the protein encoded by the wild-type sequence. In some embodiments, the at least one mutation is a loss-of-function mutation.
  • the method further comprises detecting the at least one mutation in the subject.
  • the detecting comprises subjecting a sample obtained from the subject to a sequence analysis assay.
  • the inhibitor of EZH2 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • the inhibitor of EZH2 is administered orally.
  • the inhibitor of EZH2 is formulated as a tablet.
  • the therapeutically effective amount of the inhibitor of EZH2 is between 100 mg and 3200 mg per day. -In some embodiments, the therapeutically effective amount of the inhibitor of EZH2 is 100 mg, 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, 1400 mg, 1600 mg or 3200 mg per day. In some embodiments, the therapeutically effective amount is 1600 mg per day. In some embodiments, the therapeutically effective amount of the inhibitor of is administered at 800 mg twice per day (BID).
  • BID twice per day
  • the subject or cancer expresses a wild type EZH2 protein and/or does not express a mutant EZH2 protein.
  • the subject or cancer expresses a mutant EZH2 protein.
  • the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1.
  • the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 682 of SEQ ID NO: 1.
  • the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 692 of SEQ ID NO: 1.
  • the at least one mutation consists of a single mutation. In some embodiments, the at least one mutation comprises 2 mutations or more. In some embodiments, the at least one mutation comprises 3 mutations or more. In some embodiments, the at least one mutation comprises 4 mutations or more. In some embodiments, the at least one mutation comprises S mutations or more.
  • the at least one mutation comprises 2 mutations, 3 mutations, 4 mutations, 5 mutations, 6 mutations, 7 mutations, 8 mutations, 9 mutations, 10 mutations, 11 mutations, 12 mutations, 13 mutations, 14 mutations, IS mutations, 16 mutations, 17 mutations, 18 mutations, 19 mutations, or 20 mutations.
  • the at least one mutation comprises at least one positive mutation (e.g., with or without a negative mutation). In some embodiments, the at least one mutation comprises at least one negative mutation (e.g., with or without a positive mutation). In some embodiments, the at least one mutation comprises both positive and negative mutations.
  • positive mutation refers to a mutation that sensitizes a subject, a cancer, or malignant cell or population of cells, to EZH2 treatment, or, in some embodiments, that renders a subject, cancer, or malignant cell or population of cells, more sensitive to EZH2 treatment.
  • negative mutation refers to a mutation that desensitizes a subject, a cancer, or malignant cell or population of cells, to EZH2 treatment, or, in some embodiments, that renders a subject, cancer, or malignant cell or population of cells, less sensitive to EZH2 treatment.
  • the disclosure provides a method of identifying molecular variants in tumor samples harvested from NHL patients treated with a compound of the disclosure.
  • the disclosure provides a method of identifying molecular variants in cell free circulating tumor DNA (ctDNA) harvested from NHL patients treated with a compound of the disclosure.
  • ctDNA cell free circulating tumor DNA
  • the molecular variants identified therein may correlate with clinical response, minimal residual disease or emergence of resistance.
  • Figure 1 shows an overview of the analytical and statistical methods used in this study.
  • Figures 2A-2D show somatic mutations, amplifications, and translocations detected in 92 NHL patients by best overall response.
  • Figure 2A shows all 92 NHL patients assessed.
  • Figure 2B shows NHL patients classified as responders (complete response (CR) or partial response (PR)).
  • Figure 2C shows FL patients that were classified as non-responders.
  • Figure 2D shows DLBCL patients that were classified as non-responders.
  • NHL Non-Hodgkin Lymphoma
  • CR complete response
  • PR partial response
  • SD stable disease
  • PD progressive disease
  • UK unknown response.
  • Patients with CR and PR were classified as responders.
  • Patients with SD, PD, and UK were classified as non-responders.
  • Figure 4 is a structure model of partial EZH2 protein based on the A chain of nuclear receptor binding SET domain protein 1 (NSD1). This model corresponds to amino acid residues 533-732 of EZH2 sequence of SEQ ID NO: 1. DETAILED DESCRIPTION
  • Tazemetostat demonstrates clinical activity as a monotherapy in patients with relapsed or refractory DLBCL (both GCB and non-GCB), follicular lymphoma (FL) and marginal zone lymphomas (MZL).
  • Objective responses in tumors with either wild-type or mutation in EZH2 are durable as patients are ongoing at 7+ to 21+ months.
  • Safety profile as monotherapy continues to be acceptable and favorable for combination development.
  • Baseline somatic mutation profiling revealed associations between objective response to tazemetostat and genetic alterations, e.g., mutations in genomic sequences encoding MYD88, STAT6A, SOCS1, MYC, TP53, fflSTlHlE, and histone acetyltransferases, such as, for example CREBBP and EP300.
  • EZH2 is a histone methyltransferase that is the catalytic subunit of the PRC2 complex which catalyzes the mono- through tri-methylation of lysine 27 on histone H3 (H3-K27).
  • Point mutations of the EZH2 gene at a single amino acid residue (e.g., Tyr641, herein referred to as Y641) of EZH2 have been reported to be linked to subsets of human B-cell lymphoma. Morin et al. (2010) Nat Genet 42(2): 181-5. In particular, Morin et al. reported that somatic mutations of tyrosine 641 (Y641F, Y641H, Y641N, and Y641S) of EZH2 were associated with follicular lymphoma (FL) and the germinal center B cell-like (GCB) subtype of diffuse large B-cell lymphoma (DLBCL). The mutant allele is always found associated with a wild-type allele (heterozygous) in disease cells, and the mutations were reported to ablate the enzymatic activity of the PRC2 complex for methylating an unmodified peptide substrate.
  • the mutant EZH2 refers to a mutant EZH2 polypeptide or a nucleic acid sequence encoding a mutant EZH2 polypeptide.
  • the mutant EZH2 comprises one or more mutations in its substrate pocket domain as defined in SEQ ID NO: 6.
  • the mutation may be a substitution, a point mutation, a nonsense mutation, a missense mutation, a deletion, or an insertion.
  • Exemplary substitution amino acid mutation includes a substitution at amino acid position 677, 687, 674, 685, or 641 of SEQ ID NO: 1, such as, but is not limited to a substitution of glycine (G) for the wild type residue alanine (A) at amino acid position 677 of SEQ ID NO: 1 (A677G); a substitution of valine (V) for the wild type residue alanine (A) at amino acid position 687 of SEQ ID NO: 1 (A687V); a substitution of methionine (M) for the wild type residue valine (V) at amino acid position 674 of SEQ ID NO: 1 (V674M); a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 685 of SEQ ID NO: 1 (R685H); a substitution of cysteine (C) for the wild type residue arginine (R) at amino acid position 685 of SEQ ID NO: 1 (R685C); a substitution of phen
  • the mutation may also include a substitution of serine (S) for the wild type residue asparagine (N) at amino acid position 322 of SEQ ID NO: 3 (N322S), a substitution of glutamine (Q) for the wild type residue arginine (R) at amino acid position 288 of SEQ ID NO: 3 (R288Q), a substitution of isoleucine (I) for the wild type residue threonine (T) at amino acid position 573 of SEQ ID NO: 3 (T573I), a substitution of glutamic acid (E) for the wild type residue aspartic acid (D) at amino acid position 664 of SEQ ID NO: 3 (D664E), a substitution of glutamine (Q) for the wild type residue arginine (R) at amino acid position 458 of SEQ ID NO: 5 (R458Q), a substitution of lysine (K) for the wild type residue glutamic acid (E) at amino acid position 249 of SEQ ID NO: 3 (E249
  • H694Y a substitution of alanine (A) for the wild type residue aspartic acid (D) at amino acid position 664 of SEQ ID NO: 3 (D664A), a substitution of threonine (T) for the wild type residue isoleucine (I) at amino acid position 150 of SEQ ID NO: 3 (I150T), a substitution of arginine (R) for the wild type residue isoleucine (I) at amino acid position 264 of SEQ ID NO: 3 (I264R), a substitution of leucine (L) for the wild type residue proline (P) at amino acid position 636 of SEQ ID NO: 3 (P636L), a substitution of threonine (T) for the wild type residue isoleucine (I) at amino acid position 713 of SEQ ID NO: 3 (I713T), a substitution of proline (P) for the wild type residue glutamine (Q) at amino acid position 501 of SEQ ID NO: 5 (Q501P), a substitution of
  • the mutation may be a frameshift at amino acid position 730, 391, 461, 441, 235, 254, 564, 662, 715, 405, 685, 64, 73, 656, 718, 374, 592, 505, 730, or 363 of SEQ ID NO: 3, 5 or 21 or the corresponding nucleotide position of the nucleic acid sequence encoding SEQ ID NO: 3, 5, or 21.
  • the mutation of the EZH2 may also be an insertion of a glutamic acid (E) between amino acid positions 148 and 149 of SEQ ID NO: 3, 5 or 21.
  • EZH2 mutation is a deletion of glutamic acid (E) and leucine (L) at amino acid positions 148 and 149 of SEQ ID NO: 3, 5 or 21.
  • the mutant EZH2 may further comprise a nonsense mutation at amino acid position 733, 25, 317, 62, 553, 328, 58, 207, 123, 63, 137, or 60 of SEQ ID NO: 3, 5 or 21.
  • a structure model of partial EZH2 protein based on the A chain of nuclear receptor binding SET domain protein 1 (NSD1) is provided in Figure 4. This model corresponds to amino acid residues 533-732 of EZH2 sequence of SEQ ID NO: 1.
  • the catalytic site of EZH2 is believed to reside in a conserved domain of the protein known as the SET domain.
  • the amino acid sequence of the SET domain of EZH2 is provided by the following partial sequence spanning amino acid residues 613-726 of Swiss-Prot
  • tyrosine (Y) residue shown underlined in SEQ ID NO: 7 is Tyr641 (Y641) in Swiss- Prot Accession No. Q15910 (SEQ ID NO: 1).
  • GenBank Accession No. NP_004447 spans amino acid residues 618-731 and is identical to SEQ ID NO:6.
  • the tyrosine residue corresponding to Y641 in Swiss-Prot Accession No. Q15910 shown underlined in SEQ ID NO: 7 is Tyr646 (Y646) in GenBank Accession No. NP_004447 (SEQ ID NO: 3).
  • GenBank Accession No. NP_694543 (SEQ ID NO: 5) spans amino acid residues 574-687 and is identical to SEQ ID NO: 7.
  • Y641 in Swiss-Prot Accession No. Q15910 shown underlined in SEQ ID NO: 7 is Tyr602
  • amino acid residue Y641 of human EZH2 is to be understood to refer to the tyrosine residue that is or corresponds to Y641 in Swiss-Prot
  • a Y641 mutant of human EZH2, and, equivalently, a Y641 mutant of EZH2, is to be understood to refer to a human EZH2 in which the amino acid residue corresponding to Y641 of wild-type human EZH2 is substituted by an amino acid residue other than tyrosine.
  • amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of a single amino acid residue corresponding to Y641 of wild-type human EZH2 by an amino acid residue other than tyrosine.
  • the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of phenylalanine (F) for the single amino acid residue corresponding to Y641 of wild-type human EZH2.
  • the Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641F mutant or, equivalently,
  • the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of histidine (H) for the single amino acid residue corresponding to Y641 of wild-type human EZH2.
  • H histidine
  • the Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641H mutant or, equivalently,
  • amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of asparagine (N) for the single amino acid residue corresponding to Y641 of wild-type human EZH2.
  • the Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641 N mutant or, equivalently,
  • the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of serine (S) for the single amino acid residue corresponding to Y641 of wild-type human EZH2.
  • S serine
  • the Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641S mutant or, equivalently, Y641S.
  • the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of cysteine (C) for the single amino acid residue corresponding to Y641 of wild-type human EZH2.
  • C cysteine
  • the Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641 C mutant or, equivalently, Y641C.
  • the amino acid sequence of a A677 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of a non-alanine amino acid, preferably glycine (G) for the single amino acid residue corresponding to A677 of wild- type human EZH2.
  • the A677 mutant of EZH2 according to this embodiment is referred to herein as an A677 mutant, and preferably an A677G mutant or, equivalently, A677G.
  • the amino acid sequence of a A687 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of a non-alanine amino acid, preferably valine (V) for the single amino acid residue corresponding to A687 of wild-type human EZH2.
  • the A687 mutant of EZH2 according to this embodiment is referred to herein as an A687 mutant and preferably an A687V mutant or, equivalently, A687V.
  • the amino acid sequence of a R685 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of a non-arginine amino acid, preferably histidine (H) or cysteine (C) for the single amino acid residue corresponding to R68S of wild-type human EZH2.
  • the R68S mutant of EZH2 according to this embodiment is referred to herein as an R68S mutant and preferably an R68SC mutant or an R68SH mutant or, equivalently, R685H or R685C.
  • amino acid sequence of a mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 in one or more amino acid residues in its substrate pocket domain as defined in SEQ ID NO: 6.
  • EZH2 mutant embodiment is referred to herein as an EZH2 mutant.
  • Histone acetyltransferase (HAT) enzymes of the disclosure activate gene transcription by transferring an acetyl group from acetyl CoA to form ⁇ - ⁇ -acetyllysine, which serves to modify hi stones and increase transcription by, for example, generating or exposing binding sites for protein-protein interaction domains.
  • HAT Histone acetyltransferase
  • HAT enzymes of the disclosure include, but are not limited to, those enzymes of the p300/CBP family.
  • a mutation of the disclosure may occur in a sequence encoding the p300 HAT, including the nucleotide sequence of the EP300 gene, encoding p300 (below, corresponding to GenBank Accession No. NM 001429.3, defined as Homo sapiens El A binding protein p300 (EP300), mRNA; and identified as SEQ ID NO: 19).
  • a mutation of the disclosure may occur in a sequence encoding the p300 HAT, including the amino acid sequence of the p300 protein (below, corresponding to GenBank Accession No. NP 001420.2, defined as Homo sapiens El A-binding protein, 300kD; El A-associated protein p300; p300 HAT; and identified as SEQ ID NO: 20).
  • a mutation of the disclosure may occur in a sequence encoding the CREB Binding Protein (CREBBP) HAT, including the nucleotide sequence encoding CREBBP (below, corresponding to GenBank Accession No. NM 004380, defined as Homo sapiens CREB binding protein (CREBBP), transcript variant 1, mRNA; and identified as SEQ ID NO: 1
  • a mutation of the disclosure may occur in a sequence encoding the CREB Binding Protein (CREBBP) HAT, including the amino acid sequence encoding CREBBP (below, corresponding to GenBank Accession No. NP 004371, defined as Homo sapiens CREB-binding protein isoform a; and identified as SEQ ID NO: 24).
  • CREBBP CREB Binding Protein
  • a mutation of the disclosure may occur in a sequence encoding the CREB Binding Protein (CREBBP) HAT, including the nucleotide sequence encoding CREBBP (below, corresponding to GenBank Accession No. NM OO 1079846, defined as Homo sapiens CREB binding protein (CREBBP), transcript variant 2, mRNA; and identified as SEQ ID NO: 25).
  • CREBBP CREB Binding Protein
  • the compounds of the disclosure are inhibitors of the hi stone methyltransferase EZH2 for use in the treatment of patients with non-Hodgkin lymphoma (NHL), and in patients with certain genetically defined solid tumors.
  • NHL patients non-Hodgkin lymphoma
  • Activating EZH2 mutations present in NHL patients has been implicated to predict response to EZH2 inhibition (Knutson et al., Nat. Chem. Biol. 2012; 8: 890-896, the content of which is incorporated herein by reference in its entirety).
  • a phase 1 clinical trial of tazemetostat demonstrated clinical responses in both EZH2 mutant and wild type patients (ClinicalTrials.gov identifier: NCT01897571).
  • the present disclosure provides a multi-gene NHL targeted next generation sequencing (NGS) panel (e.g., a 39-gene panel or a 62-gene panel, or a panel combining a plurality of genes or gene products referred to herein) capable of analyzing samples from malignant cells, tissues, or body fluids, e.g., archive tissue or cell-free circulating tumor DNA (ctDNA) isolated from plasma.
  • NGS next generation sequencing
  • the NGS panel is capable of identifying molecular variants, including specific somatic sequence mutations (single base and
  • insertion/deletion e.g., EZH2
  • amplifications e.g., BLC2
  • translocations e.g., BCL2 and MYC
  • sequencing of phase 1 NHL patients utilizing a 62 gene NHL NGS panel revealed a complex genetic landscape with epigenetic modifiers CREBBP and KMT2D representing the most frequently mutated genes in this sample set. Further aspects of the disclosure provide for an NGS panel with the ability to determine molecular profiles using ctDNA that enables patient
  • profiling ctDNA enables longitudinal monitoring of a patient's mutation burden without the need for tumor biopsies.
  • mutations identified by the NGS panel disclosed herein may be used for patient stratification. Accordingly, in some embodiments, the disclosure provides a method of selecting a patient for cancer treatment if the patient has one or more mutations disclosed herein. In some embodiments, the patient selected for the cancer treatment has two or more (e.g., two, three, four, five, six, seven, eight, or more) mutations disclosed herein.
  • a method in which a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of one or more mutations associated with a positive response to such treatment in the subject, e.g., as determined by ctDNA analysis.
  • a mutation (or a combination of two or more mutations) associated with a positive response is a mutation (or a combination of mutations) that is present only in patients who responded with complete or partial response or, in some embodiments, with stable disease in any of the studies presented herein, e.g., those summarized in Figures 1-3.
  • a mutation (or a combination of two or more mutations) associated with a positive response is a mutation (or a combination of mutations) that is not randomly distributed within the patient population examined, but is overrepresented in those patients who responded with a complete or partial response or, in some embodiments, stable disease, in any of the studies presented herein, e.g., those summarized in Figures 1-3.
  • a mutation (or combination of mutations) associated with a positive response is a mutation (or combination of mutations) that is overrepresented in the responding (CR, PR, or, in some embodiments, SD) patient population at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, or at least 10-fold, as compared to the patient population that did not respond or responded with progressive disease (PD).
  • PD progressive disease
  • a method in which a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the absence of one or more mutations associated with a negative response to such treatment in the subject, e.g., as determined by ctDNA analysis.
  • a mutation (or a combination of two or more mutations) associated with a negative response is a mutation (or a combination of mutations) that is present only in patients who did not respond or responded with progressive disease (PD) in any of the studies presented herein, e.g., those summarized in Figures 1-3.
  • a mutation (or a combination of two or more mutations) associated with a negative response is a mutation (or a combination of mutations) that is not randomly distributed within the patient population examined, but is overrepresented in those patients who did not respond or responded with progressive disease in any of the studies presented herein, e.g., those summarized in Figures 1-3.
  • a mutation (or combination of mutations) associated with a negative response is a mutation (or combination of mutations) that is overrepresented in the non-responding or progressive disease (PD) patient population at least 2-fold, at least 3-fold, at least 4-fold, at least S-fold, or at least 10-fold, as compared to the patient population that responded with CR, PR, or, in some embodiments, SD.
  • PD non-responding or progressive disease
  • a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of two or more (e.g., two, three, four, five, six, seven, eight, or more) mutations in the subject that match the mutations observed in a profile of a patient who exhibited a complete or partial response in any of the studies described herein (e.g., those summarized in Figures 1-3).
  • an EZH2 inhibitor e.g., an EZH2 inhibitor disclosed herein
  • a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of a mutation profile (e.g., of two or more (e.g., two, three, four, five, six, seven, eight, or more)) mutations in the subject that match the mutation profile of a patient who exhibited a complete or partial response in any of the studies described herein (e.g., those summarized in Figures 1-3).
  • a mutation profile e.g., of two or more (e.g., two, three, four, five, six, seven, eight, or more) mutations in the subject that match the mutation profile of a patient who exhibited a complete or partial response in any of the studies described herein (e.g., those summarized in Figures 1-3).
  • a mutation in a gene or gene product is detected by comparing a given sequence with a reference sequence, e.g., a human reference genome sequence (e.g., human reference genome hgl9), and identifying a mismatch in the sequence at hand as compared to the reference sequence.
  • a reference sequence e.g., a human reference genome sequence (e.g., human reference genome hgl9)
  • a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of two or more (e.g., two, three, four, five, six, seven, eight, or more) mutations in the subject that match the mutations observed in a profile of a patient who exhibited stable disease in any of the studies described herein (e.g., those summarized in Figures 1-3).
  • an EZH2 inhibitor e.g., an EZH2 inhibitor disclosed herein
  • a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of a mutation profile (e.g., two or more (e.g., two, three, four, five, six, seven, eight, or more)) mutations in the subject that match the mutation profile of a patient who exhibited stable disease in any of the studies described herein (e.g., those summarized in Figures 1-3).
  • a mutation profile e.g., two or more (e.g., two, three, four, five, six, seven, eight, or more) mutations in the subject that match the mutation profile of a patient who exhibited stable disease in any of the studies described herein (e.g., those summarized in Figures 1-3).
  • methods of treating cancer is provided that comprises
  • a therapeutically effective amount of an inhibitor of EZH2 to a subject in need thereof, wherein the subject has at least one mutation in one or more sequences encoding a gene or a gene product (e.g., a transcript, mRNA, or protein) listed in Tables 1-5, and/or Figures 1-3.
  • a gene or a gene product e.g., a transcript, mRNA, or protein listed in Tables 1-5, and/or Figures 1-3.
  • the subject has at least one mutation in in one or more sequences encoding: MYD88, STAT6A, SOCS1, MYC, HIST1H1E, ABL1, ACVR1, AKT1, AKT2, ALK, APC, AR, ARID1A, ARID1B, ASXL1, ATM, ATRX, AURKA, AXIN2, BAP1, BCL2, BCR, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRIP1, BTK, BUB IB, CALR, CBL, CCND1, CCNE1, CDC73, CDH1, CDK4, CDK6, CDKN1B, CDKN2A, CDKN2B, CDKN2C, CEBPA, CHEK2, CIC, CREBBP, CSF1R, CTNNB1, CYLD, DAXX, DDB2, DDR2, DICERl, DNMT3A, EGFR, EP300, ERBB2, ERBB3, ERBB4,
  • the subject has at least one mutation in one or more sequences encoding ABLl, ACVR1, AKT1, AKT2, ALK, APC, AR, ARIDIA, ARID IB, ASXL1, ATM, ATRX, AURKA, AXIN2, BAP1, BCL2, BCR, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRIP1, BTK, BUB IB, CALR, CBL, CCND1, CCNE1, CDC73, CDH1, CDK4, CDK6, CDKN1B, CDKN2A, CDKN2B, CDKN2C, CEBPA, CHEK2, CIC, CREBBP, CSF1R, CTNNB1, CYLD, DAXX, DDB2, DDR2, DICERl, DNMT3A, EGFR, EP300, ERBB2, ERBB3, ERBB4, ERCC1, ERCC2, ERCC3, ERCC4, ERCC5, ESR1, ETVl,
  • the subject has at least one mutation in one or more sequences encoding ARID1 A, ATM, B2M, BCL2, BCL6, BCL7A, BRAF, BTG1, CARD11, CCND3, CD58, CD79B, CDKN2A, CREBBP, EP300, EZH2, FOXOl, GNA13, fflSTlHIB, fflSTlHIC, HIST1H1E, IKZF3, IRF4, ⁇ , KDM6A, KIT, KMT2D, KRAS, MEF2B, MYC, MYD88, NOTCH1, NOTCH2, NRAS, PIK3CA, PIM1, POU2F2, PRDM1, PTEN, PTPN1, PTPN11, PTPN6, PTPRD, RBI, S1PR2, SGK1, SMARCB1, SOCS1, STAT6, TBL1XR1, TNFAIP3, TNFRSF14, TP53, XPOl.
  • the subject has at least one mutation in one or more sequences encoding AKTl, ALK, ARID 1 A, ATM, B2M, BCL2, BCL6, BCL7A, BTG2, CARDll, CCND3, CD79B, CDKN2A, CREBBP, EP300, EZH2, FBXW7, FOXOl, HLA-C, HRAS, IKZF3, IRF4, KDM6A, KRAS, MEF2B, MYD88, NOTCH1, NPM1, NRAS, PIK3CA, PIM1, PRDMl, PTEN, RBI, RBBP4,
  • the subject has at least one mutation in one or more sequences encoding ALK, EWSR1, ROS1, BCL2, MLL,
  • the subject has at least one mutation in one or more sequences encoding ALK (Intron 19), BCL2 (MBR breakpoint region), BCL2 (MCR breakpoint region), BCL6, CD274, CIITA, MYC (entire Gene + 40kbp upstream), and/or PDCD1 LG2.
  • the subject has at least one mutation in one or more sequences encoding BCL2, CD274 (PDL1), FOXP1, JAK2, KDM4C, PDCD1LG2 (PDL2), and/or REL. In some embodiments, the subject has at least one mutation in one or more sequences encoding ARIDl A, ATM, B2M, BCL2, BCL6, BCL7A, BRAF,
  • the subject has at least one mutation in one or more sequences encoding
  • ARIDl A B2M, BCL2, BCL6, CARDl 1, CCND3, CD274 (PDL1), CD58, CD79B, CDKN2A, CREBBP, EZH2, EP300, FOXOl, GNA13, HIST1H1B, fflSTlHIC, fflSTlHlE, KMT2D, KRAS, MEF2B, MYC, MYD88 (273P), PDCD1LG2 (PDL2), PIM1, POU2F2, PRDMl, SOCS1, STAT6, TNFAIP3, and/or TNFRSF14.
  • PDL1LG2 PIM1, POU2F2
  • PRDMl SOCS1, STAT6, TNFAIP3, and/or TNFRSF14.
  • the subject has at least one mutation in in one or more sequences encoding: EZH2, MYD88, STAT6A, SOCS1, MYC, and/or fflSTlHlE, [070J
  • the subject has at least one mutation that decreases or abolishes the function of a gene product (e.g., a transcript, mRNA, or protein) encoded by the mutated sequence as compared to the function of the respective gene product encoded by the wild-type sequence.
  • a gene product e.g., a transcript, mRNA, or protein
  • Such mutations are also sometimes referred to as loss-of-function mutations.
  • Many loss-of-function mutations for the genes and gene products referred to herein that are suitable for some embodiments of this disclosure will be known to the skilled artisan.
  • the subject has a loss-of-function mutation in SOCS1.
  • the subject has at least one mutation that increases the function of a gene product (e.g., a transcript, mRNA, or protein) encoded by the mutated sequence as compared to the function of the respective gene product encoded by the wild-type sequence.
  • a gene product e.g., a transcript, mRNA, or protein
  • Such mutations are also sometimes referred to as gain-of-function mutations or activating mutations.
  • gain-of- function mutations for the genes and gene products referred to herein that are suitable for some embodiments of this disclosure will be known to the skilled artisan.
  • the subject has a gain-of-function mutation in a sequence encoding EZH2, MYD88, STAT6, or MYC.
  • the subject has at least one loss-of-function and at least one gain-of function mutation.
  • the subject has at least one gain-of-function mutation in a sequence encoding EZH2 or STAT6, and at least one loss-of-function mutation in a sequence encoding SOCS1.
  • the subject does not have a specific mutation, e.g., a gain-of-function in a sequence encoding MYC or a loss-of-function mutation in SOCS1.
  • the subject expresses a mutant EZH2 protein.
  • the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1, a substitution of any amino acid other than alanine (A) for alanine (A) at position 682 of SEQ ID NO: 1, and/or a substitution of any amino acid other than alanine (A) for alanine (A) at position 692 of SEQ ID NO: 1.
  • the subject expresses at least one mutant MYD88, STAT6, and/or a SOCS1 protein, either in addition to the mutant EZH2 protein or in the absence of a mutant EZH2 protein. In some embodiments, the subject does not express a mutant MYC and/or a mutant HIST1H1E protein. In some embodiments, the mutant EZH2 protein, the mutant MYD88 protein, the mutant STAT6 protein, and/or the mutant MYC protein exhibits an increase in activity as compared to the respective wild-type protein. In some embodiments, the mutant SOCS1 protein exhibits a decreased activity as compared to the respective wild-type SOCS1 protein.
  • the methods provided herein further comprise detecting the at least one mutation in the subject.
  • detecting may, in some embodiments, comprise subjecting a sample obtained from the subject to a suitable sequence analysis assay, e.g., to a next generation sequencing assay.
  • suitable sequencing assays are provided herein or otherwise known to those of skill in the art, and the disclosure is not limited in this respect.
  • Some aspects of this disclosure provide methods comprising selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of at least one mutation associated with a positive response to such treatment in the subject and/or based on the absence of at least one mutation associated with no response or with a negative response to such treatment in the subject.
  • the at least one mutation associated with a positive response comprises (a) an EZH2 mutation (e.g., a gain-of-function EZH2 mutation); (b) a histone acetyl transferase (HAT) mutation; (c) a STAT6 mutation (e.g., a gain-of-function STAT6 mutation); (d) a MYD88 mutation (e.g., a gain-of-function MYD88 mutation); and/or (e) a SOCS1 mutation (e.g., a loss-of-function SOCS1 mutation).
  • an EZH2 mutation e.g., a gain-of-function EZH2 mutation
  • HAT histone acetyl transferase
  • STAT6 mutation e.g., a gain-of-function STAT6 mutation
  • MYD88 mutation e.g., a gain-of-function MYD88 mutation
  • SOCS1 mutation e.g., a loss-of
  • the at least one mutation associated with no response or with a negative response comprises (a) a MYC mutation (e.g., a gain-of-function MYC mutation); and/or (b) a HISTIHIE mutation.
  • the method comprises detecting the at least one mutation associated with a positive response and/or the at least one mutation associated with no response or a negative response in a sample obtained from the subject by subjecting the sample to a suitable sequence analysis assay.
  • the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having at least one of a MYD88 mutation, a STAT6A mutation, and a SOCS1 mutation, and/or (b) not having at least one of a MYC mutation and/or a HISTIHIE mutation.
  • the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having at least one of a MYD88 mutation, a STAT6A mutation, and a SOCS1 mutation, and (b) not having a MYC mutation and a HISTIHIE mutation.
  • Some aspects of this disclosure provide methods for selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of a mutation profile in the subject that matches a mutation profile (e.g., at least 2, at least 3, at least 4, or at least S, or more mutations, or, in some embodiments, all mutations), of a patient exhibiting a complete or partial response or stable disease as described in any of Figures 1-3.
  • a mutation profile e.g., at least 2, at least 3, at least 4, or at least S, or more mutations, or, in some embodiments, all mutations
  • a "normal" cell may be used as a basis of comparison for one or more characteristics of a cancer cell, including the presence of one or more mutations in a hi stone acetyltransferase that result in a decreased activity of the enzyme.
  • the one or more mutations in a histone acetyltransferase may result in a decreased acetylation activity or efficacy of the enzyme, and, consequently, a reduced or decreased level of acetylation of at least one lysine on Histone 3 (H3).
  • the one or more mutations in a histone acetyltransferase may result in a decreased acetylation activity or efficacy of the enzyme, and, consequently, a reduced or decreased level of acetylation of lysine 27 on Histone 3 (H3) (H3K27).
  • a "normal cell” is a cell that cannot be classified as part of a "cell proliferative disorder”. A normal cell lacks unregulated or abnormal growth, or both, that can lead to the development of an unwanted condition or disease.
  • a normal cell expresses a comparable amount of EZH2 as a cancer cell.
  • a normal cell contains a wild type sequence for all histone acetyltransferases, expresses a histone
  • acetyltransferase transcript without mutations, and expresses a histone acetyltransferase protein without mutations that retains all functions a normal activity levels.
  • contacting a cell refers to a condition in which a compound or other composition of matter is in direct contact with a cell, or is close enough to induce a desired biological effect in a cell.
  • treating or “treat” describes the management and care of a subject for the purpose of combating a disease, condition, or disorder and includes the administration of an EZH2 inhibitor of the disclosure, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph or solvate thereof, to alleviate the symptoms or complications of cancer or to eliminate the cancer.
  • the term "alleviate” is meant to describe a process by which the severity of a sign or symptom of cancer is decreased.
  • a sign or symptom can be alleviated without being eliminated.
  • the administration of pharmaceutical compositions of the disclosure leads to the elimination of a sign or symptom, however, elimination is not required.
  • Effective dosages are expected to decrease the severity of a sign or symptom. For instance, a sign or symptom of a disorder such as cancer, which can occur in multiple locations, is alleviated if the severity of the cancer is decreased within at least one of multiple locations.
  • severity is meant to describe the potential of cancer to transform from a precancerous, or benign, state into a malignant state. Alternatively, or in addition, severity is meant to describe a cancer stage, for example, according to the TNM system
  • Cancer stage refers to the extent or severity of the cancer, based on factors such as the location of the primary tumor, tumor size, number of tumors, and lymph node involvement (spread of cancer into lymph nodes).
  • Tumor grade is a system used to classify cancer cells in terms of how abnormal they look under a microscope and how quickly the tumor is likely to grow and spread. Many factors are considered when determining tumor grade, including the structure and growth pattern of the cells. The specific factors used to determine tumor grade vary with each type of cancer. Severity also describes a histologic grade, also called differentiation, which refers to how much the tumor cells resemble normal cells of the same tissue type (see, National Cancer Institute, www.cancer.gov). Furthermore, severity describes a nuclear grade, which refers to the size and shape of the nucleus in tumor cells and the percentage of tumor cells that are dividing (see, National Cancer Institute, www. cancer.gov).
  • severity describes the degree to which a tumor has secreted growth factors, degraded the extracellular matrix, become vascularized, lost adhesion to juxtaposed tissues, or metastasized. Moreover, severity describes the number of locations to which a primary tumor has metastasized. Finally, severity includes the difficulty of treating tumors of varying types and locations. For example, inoperable tumors, those cancers which have greater access to multiple body systems (hematological and immunological tumors), and those which are the most resistant to traditional treatments are considered most severe.
  • symptom is defined as an indication of disease, illness, injury, or that something is not right in the body. Symptoms are felt or noticed by the individual experiencing the symptom, but may not easily be noticed by others. Others are defined as non- health-care professionals.
  • signs are also defined as an indication that something is not right in the body. But signs are defined as things that can be seen by a doctor, nurse, or other health care professional.
  • Cancer is a group of diseases that may cause almost any sign or symptom. The signs and symptoms will depend on where the cancer is, the size of the cancer, and how much it affects the nearby organs or structures. If a cancer spreads (metastasizes), then symptoms may appear in different parts of the body.
  • Cancer may also cause symptoms such as fever, fatigue, or weight loss. This may be because cancer cells use up much of the body's energy supply or release substances that change the body's metabolism. Or the cancer may cause the immune system to react in ways that produce these symptoms. While the signs and symptoms listed above are the more common ones seen with cancer, there are many others that are less common and are not listed here. However, all art-recognized signs and symptoms of cancer are contemplated and encompassed by the disclosure.
  • Treating cancer may result in a reduction in size of a tumor.
  • a reduction in size of a tumor may also be referred to as "tumor regression".
  • tumor size is reduced by 5% or greater relative to its size prior to treatment; more preferably, tumor size is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75% or greater.
  • Size of a tumor may be measured by any reproducible means of measurement. The size of a tumor may be measured as a diameter of the tumor.
  • Treating cancer may result in a reduction in tumor volume.
  • tumor volume is reduced by 5% or greater relative to its size prior to treatment; more preferably, tumor volume is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75% or greater.
  • Tumor volume may be measured by any reproducible means of measurement.
  • Treating cancer may result in a decrease in number of tumors.
  • tumor number is reduced by 5% or greater relative to number prior to treatment; more preferably, tumor number is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75%.
  • Number of tumors may be measured by any reproducible means of measurement.
  • the number of tumors may be measured by counting tumors visible to the naked eye or at a specified magnification.
  • the specified magnification is 2x, 3x, 4x, 5x, 10x, or 50x.
  • Treating cancer may result in a decrease in number of metastatic lesions in other tissues or organs distant from the primary tumor site.
  • the number of metastatic lesions is reduced by 5% or greater relative to number prior to treatment; more preferably, the number of metastatic lesions is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75%.
  • the number of metastatic lesions may be measured by any reproducible means of measurement.
  • the number of metastatic lesions may be measured by counting metastatic lesions visible to the naked eye or at a specified magnification.
  • the specified magnification is 2x, 3x, 4x, 5x, lOx, or 50x.
  • an effective amount of an EZH2 inhibitor of the disclosure is not significantly cytotoxic to normal cells.
  • a therapeutically effective amount of an EZH2 inhibitor of the disclosure is not significantly cytotoxic to normal cells if administration of the EZH2 inhibitor of the disclosure in a therapeutically effective amount does not induce cell death in greater than 10% of normal cells.
  • a therapeutically effective amount of an EZH2 inhibitor of the disclosure does not significantly affect the viability of normal cells if administration of the compound in a therapeutically effective amount does not induce cell death in greater than 10% of normal cells.
  • [092J EZH2 inhibitors of the disclosure comprise tazemetostat (EPZ-6438):
  • Tazemetostat is also described in US Patent Nos. 8,410,088, 8,765,732, and 9,090,562 (the contents of which are each incorporated herein in their entireties).
  • Tazemetostat or a pharmaceutically acceptable salt thereof, as described herein, is potent in targeting both WT and mutant EZH2.
  • Tazemetostat is orally bioavailable and has high selectivity to EZH2 compared with other histone methyltransferases (i.e., >20,000 fold selectivity by Ki).
  • tazemetostat has targeted methyl mark inhibition that results in the killing of genetically defined cancer cells in vitro. Animal models have also shown sustained in vivo efficacy following inhibition of the target methyl mark. Clinical trial results described herein also demonstrate the safety and efficacy of tazemetostat.
  • tazemetostat or a pharmaceutically acceptable salt thereof is administered to the subject at a dose of approximately 100 mg to approximately 3200 mg daily, such as about 100 mg BID to about 1600 mg BID (e.g., 100 mg BID, 200 mg BID, 400 mg BID, 800 mg BID, or 1600 mg BID), for treating a NHL. On one embodiment the dose is 800 mg BID.
  • EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of:
  • EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of Compound E:
  • EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of GSK-126, having the following formula:
  • EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of Compound F:
  • EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of any one of Compounds Ga-Gc:
  • EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of CPI-1205 or GSK343.
  • the EZH2 inhibitor is an EZH2 inhibitor described in US 8,536,179 (describing GSK-126 among other compounds and corresponding to WO 2011/140324), the entire contents of each of which are incorporated herein by reference.
  • the EZH2 inhibitor is an EZH2 inhibitor described in PCT/US2014/015706, published as WO 2014/124418, in PCT/US2013/025639, published as WO 2013/120104, and in US 14/839,273, published as US 2015/0368229, the entire contents of each of which are incorporated herein by reference.
  • the compound disclosed herein is the compound itself, i.e., the free base or "naked" molecule.
  • the compound is a salt thereof, e.g., a mono-HCl or tri-HCl salt, mono-HBr or tri-HBr salt of the naked molecule.
  • N-oxides can be converted to N-oxides by treatment with an oxidizing agent (e.g., 3-chloroperoxybenzoic acid (/HCPBA) and/or hydrogen peroxides) to afford other compounds suitable for any methods disclosed herein.
  • an oxidizing agent e.g., 3-chloroperoxybenzoic acid (/HCPBA) and/or hydrogen peroxides
  • all shown and claimed nitrogen-containing compounds are considered, when allowed by valency and structure, to include both the compound as shown and its N-oxide derivative (which can be designated as N DO or N + -0 " ).
  • the nitrogens in the compounds disclosed herein can be converted to N-hydroxy or N-alkoxy compounds.
  • N- hydroxy compounds can be prepared by oxidation of the parent amine by an oxidizing agent such as /H-CPBA.
  • nitrogen-containing compounds are also considered, when allowed by valency and structure, to cover both the compound as shown and its N- hydroxy (i.e., N-OH) and N-alkoxy (i.e., N-OR, wherein R is substituted or unsubstituted Ci-C 6 alkyl, Ci-Ce alkenyl, C1-C6 alkynyl, 3-14-membered carbocycle or 3-14-membered heterocycle) derivatives.
  • N-OH N-hydroxy
  • N-alkoxy i.e., N-OR, wherein R is substituted or unsubstituted Ci-C 6 alkyl, Ci-Ce alkenyl, C1-C6 alkynyl, 3-14-membered carbocycle or 3-14-membered heterocycle
  • Racemic mixture means compounds that have identical molecular formulae but differ in the sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers.” Stereoisomers that are not mirror images of one another are termed “diastereoisomers,” and stereoisomers that are non-superimposable mirror images of each other are termed “enantiomers” or sometimes optical isomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a "racemic mixture.”
  • Chiral isomer means a compound with at least one chiral center. Compounds with more than one chiral center may exist either as an individual diastereomer or as a mixture of diastereomers, termed "diastereomeric mixture.” When one chiral center is present, a stereoisomer may be characterized by the absolute configuration (R or S) of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al., Angew. Chem. Inter. Edit.
  • Gaometric isomer means the diastereomers that owe their existence to hindered rotation about double bonds or a cycloalkyl linker (e.g., 1, 3-cylcobutyl). These configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold-Prelog rules.
  • Tautomer is one of two or more structural isomers that exist in equilibrium and is readily converted from one isomeric form to another. This conversion results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. Tautomers exist as a mixture of a tautomeric set in solution. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent and pH. The concept of tautomers that are interconvertible by tautomerization is called tautomerism.
  • keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs.
  • Ring- chain tautomerism arises as a result of the aldehyde group (-CHO) in a sugar chain molecule reacting with one of the hydroxy groups (-OH) in the same molecule to give it a cyclic (ring- shaped) form as exhibited by glucose.
  • keto-enol equilibria is between pyridin-2(lH)-ones and the corresponding pyridin-2-ols, as shown below.
  • the compounds disclosed herein may be depicted as different tautomers. It should also be understood that when compounds have tautomeric forms, all tautomeric forms are intended to be included in the scope of the disclosure, and the naming of the compounds does not exclude any tautomer form.
  • the compounds disclosed herein include the compounds themselves, as well as their salts and their solvates, if applicable.
  • a salt for example, can be formed between an anion and a positively charged group (e.g., amino) on an aryl- or heteroaryl-substituted benzene compound.
  • Suitable anions include chloride, bromide, iodide, sulfate, bi sulfate, sulfamate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, glutamate, glucuronate, glutarate, malate, maleate, succinate, fumarate, tartrate, tosylate, salicylate, lactate, naphthalenesulfonate, and acetate (e.g., trifluoroacetate).
  • pharmaceutically acceptable anion refers to an anion suitable for forming a pharmaceutically acceptable salt.
  • a salt can also be formed between a cation and a negatively charged group (e.g., carboxylate) on an aryl- or heteroaryl-substituted benzene compound.
  • Suitable cations include sodium ion, potassium ion, magnesium ion, calcium ion, and an ammonium cation such as tetramethylammonium ion.
  • the aryl- or heteroaryl-substituted benzene compounds also include those salts containing quaternary nitrogen atoms.
  • the ratio of the compound to the cation or anion of the salt can be 1:1, or any ration other than 1:1, e.g., 3:1, 2:1, 1:2, or 1:3.
  • the compounds disclosed herein for example, the salts of the compounds, can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules.
  • Nonlimiting examples of hydrates include monohydrates, dihydrates, etc.
  • Nonlimiting examples of solvates include ethanol solvates, acetone solvates, etc.
  • Solvate means solvent addition forms that contain either stoichiometric or non- stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate; and if the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one molecule of the substance in which the water retains its molecular state as EbO.
  • analog refers to a chemical compound that is structurally similar to another but differs slightly in composition (as in the replacement of one atom by an atom of a different element or in the presence of a particular functional group, or the
  • an analog is a compound that is similar or comparable in function and appearance, but not in structure or origin to the reference compound.
  • the term “derivative” refers to compounds that have a common core structure, and are substituted with various groups as described herein.
  • all of the compounds represented by Formula (I) are aryl- or heteroaryl-substituted benzene compounds, and have Formula (I) as a common core.
  • bioisostere refers to a compound resulting from the exchange of an atom or of a group of atoms with another, broadly similar, atom or group of atoms.
  • the objective of a bioisosteric replacement is to create a new compound with similar biological properties to the parent compound.
  • the bioisosteric replacement may be physicochemically or topological ⁇ based.
  • Examples of carboxylic acid bioisosteres include, but are not limited to, acyl
  • isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium
  • isotopes of carbon include C-13 and C-14.
  • compositions comprising at least one EZH2 inhibitor described herein in combination with at least one pharmaceutically acceptable excipient or carrier.
  • a "pharmaceutical composition” is a formulation containing the EZH2 inhibitors of the present disclosure in a form suitable for administration to a subject.
  • the pharmaceutical composition is in bulk or in unit dosage form.
  • the unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler or a vial.
  • the quantity of active ingredient ⁇ e.g., a formulation of the disclosed compound or salt, hydrate, solvate or isomer thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved.
  • active ingredient ⁇ e.g., a formulation of the disclosed compound or salt, hydrate, solvate or isomer thereof
  • the dosage will also depend on the route of administration.
  • routes of administration A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like.
  • Dosage forms for the topical or transdermal administration of a compound of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers or propellants that are required.
  • the phrase "pharmaceutically acceptable” refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable excipient” means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use.
  • a “pharmaceutically acceptable excipient” as used in the disclosure includes both one and more than one such excipient.
  • a pharmaceutical composition of the disclosure is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), and transmucosal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens;
  • antioxidants such as ascorbic acid or sodium bisulfite
  • chelating agents such as
  • ethylenediaminetetraacetic acid ethylenediaminetetraacetic acid
  • buffers such as acetates, citrates or phosphates
  • agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • a compound or pharmaceutical composition of the disclosure can be administered to a subject in many of the well-known methods currently used for chemotherapeutic treatment.
  • a compound of the disclosure may be injected directly into tumors, injected into the blood stream or body cavities or taken orally or applied through the skin with patches.
  • the dose chosen should be sufficient to constitute effective treatment but not as high as to cause unacceptable side effects.
  • the state of the disease condition e.g., cancer, precancer, and the like
  • the health of the patient should preferably be closely monitored during and for a reasonable period after treatment.
  • terapéuticaally effective amount refers to an amount of an EZH2 inhibitor, composition, or pharmaceutical composition thereof effective to treat, ameliorate, or prevent an identified disease or condition, or to exhibit a detectable therapeutic or inhibitory effect.
  • the effect can be detected by any assay method known in the art.
  • the precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration.
  • Therapeutically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician.
  • the disease or condition to be treated is cancer, including but not limited to, B cell lymphoma, including activated B-cell (ABC) and germinal B-cell (GBC) subtypes.
  • the therapeutically effective amount can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models, usually rats, mice, rabbits, dogs, or pigs.
  • the animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • Therapeutic/prophylactic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., EDso (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LDso/EDso.
  • Pharmaceutical compositions that exhibit large therapeutic indices are preferred.
  • the dosage may vary within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
  • Dosage and administration are adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect.
  • Factors which may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy.
  • Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular formulation.
  • compositions containing an EZH2 inhibitor of the present disclosure may be manufactured in a manner that is generally known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes.
  • Pharmaceutical compositions may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and/or auxiliaries that facilitate processing of the active compounds into preparations that can be used pharmaceutically. Of course, the appropriate formulation is dependent upon the route of administration chosen.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible pharmaceutically acceptable carrier.
  • compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
  • Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser, which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Sy stemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the active compounds e.g., EZH2 inhibitors of the disclosure
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the disclosure are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.
  • the dosages of the pharmaceutical compositions used in accordance with the disclosure vary depending on the agent, the age, weight, and clinical condition of the recipient patient, and the experience and judgment of the clinician or practitioner administering the therapy, among other factors affecting the selected dosage.
  • the dose should be sufficient to result in slowing, and preferably regressing, the growth of the tumors and also preferably causing complete regression of the cancer.
  • An effective amount of a pharmaceutical agent is that which provides an objectively identifiable improvement as noted by the clinician or other qualified observer. For example, regression of a tumor in a patient may be measured with reference to the diameter of a tumor. Decrease in the diameter of a tumor indicates regression. Regression is also indicated by failure of tumors to reoccur after treatment has stopped.
  • the term "dosage effective manner" refers to amount of an active compound to produce the desired biological effect in a subject or cell.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • pharmaceutically acceptable salts refer to derivatives of the compounds of the present disclosure wherein the parent compound is modified by making acid or base salts thereof.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines, alkali or organic salts of acidic residues such as carboxylic acids, and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include, but are not limited to, those derived from inorganic and organic acids selected from 2-acetoxybenzoic, 2-hydroxyethane sulfonic, acetic, ascorbic, benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edetic, ethane disulfonic, 1,2-ethane sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, glycollyarsanilic,
  • hexylresorcinic hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxymaleic, hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic, malic, mandelic, methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic, phenylacetic, phosphoric, polygalacturonic, propionic, salicyclic, stearic, sub acetic, succinic, sulfamic, sulfanilic, sulfuric, tannic, tartaric, toluene sulfonic, and the commonly occurring amine acids, e.g., glycine, alanine, phenylalanine, arginine, etc.
  • amine acids e.g., glycine, alanine, phenylalanine, arg
  • compositions include hexanoic acid, cyclopentane propionic acid, pyruvic acid, malonic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo-[2.2.2]-oct-2-ene-l-carboxylic acid, 3- phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, muconic acid, and the like.
  • the present disclosure also encompasses salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine,
  • the EZH2 inhibitors of the present disclosure can also be prepared as esters, for example, pharmaceutically acceptable esters.
  • a carboxylic acid function group in a compound can be converted to its corresponding ester, e.g., a methyl, ethyl or other ester.
  • an alcohol group in a compound can be converted to its corresponding ester, e.g., an acetate, propionate or other ester.
  • the EZH2 inhibitors of the present disclosure can also be prepared as prodrugs, for example, pharmaceutically acceptable prodrugs.
  • pro-drug and “prodrug” are used interchangeably herein and refer to any compound which releases an active parent drug in vivo. Since prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, etc.), the compounds of the present disclosure can be delivered in prodrug form. Thus, the present disclosure is intended to cover prodrugs of the presently claimed compounds, methods of delivering the same and compositions containing the same.
  • Prodrugs are intended to include any covalently bonded carriers that release an active parent drug of the present disclosure in vivo when such prodrug is administered to a subject.
  • Prodrugs in the present disclosure are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
  • Prodrugs include compounds of the present disclosure wherein a hydroxy, amino, sulfhydryl, carboxy or carbonyl group is bonded to any group that may be cleaved in vivo to form a free hydroxyl, free amino, free sulfhydryl, free carboxy or free carbonyl group, respectively.
  • prodrugs include, but are not limited to, esters (e.g., acetate,
  • dialkylaminoacetates formates, phosphates, sulfates and benzoate derivatives
  • carbamates e.g., N,N-dimethylaminocarbonyl
  • esters e.g., ethyl esters, morpholinoethanol esters
  • N-acyl derivatives e.g., N-acetyl
  • Mannich bases Schiff bases and enaminones of amino functional groups
  • oximes acetals, ketals and enol esters of ketone and aldehyde functional groups in compounds of the disclosure, and the like, See Bundegaard, H., Design of Prodrugs, pl-92, Elesevier, New York-Oxford (1985).
  • EZH2 inhibitors or pharmaceutically acceptable salts, esters or prodrugs thereof, are administered orally, nasally, transdermally, pulmonary, inhalationally, buccally,
  • the compound is administered orally.
  • One skilled in the art will recognize the advantages of certain routes of administration.
  • the dosage regimen utilizing the compounds is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed.
  • An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
  • the dosage regimen can be daily administration (e.g., every 24 hours) of a compound of the present disclosure.
  • the dosage regimen can be daily administration for consecutive days, for example, at least two, at least three, at least four, at least five, at least six or at least seven consecutive days. Dosing can be more than one time daily, for example, twice, three times or four times daily (per a 24 hour period).
  • the dosing regimen can be a daily administration followed by at least one day, at least two days, at least three days, at least four days, at least five days, or at least six days, without administration.
  • Suitable pharmaceutically acceptable carriers include inert solid fillers or diluents and sterile aqueous or organic solutions.
  • the compounds will be present in such pharmaceutical compositions in amounts sufficient to provide the desired dosage amount in the range described herein.
  • Methods of the disclosure for treating cancer including treating a B cell lymphoma, including the activated B-cell (ABC) and germinal B-cell (GBC) subtypes.
  • methods of the disclosure are used to treat a subject having a B cell lymphoma.
  • the B cell lymphoma cell and/or the subject are characterized as having one or more mutations in a sequence that encodes a histone acetyltransferase (HAT).
  • B cell lymphoma cells may contain a mutation in a gene that encodes a HAT, a corresponding HAT transcript (or cDNA copy thereof), or a HAT protein that decreases/inhibits an activity of a HAT protein.
  • the mutation in a gene that encodes a HAT, a corresponding HAT transcript (or cDNA copy thereof), or a HAT protein that decreases/inhibits an activity of a HAT protein decreases or inhibits an acetylation activity or efficacy of the enzyme, resulting in a decreased level of acetylation of one or more lysines of histone 3 (H3) (e.g., H3K27).
  • H3 histone 3
  • the presence of the HAT mutation resulting in a decreased level of acetylation of one or more lysines of histone 3 (H3) (e.g., H3K27) in a cell renders that cell sensitive to oncogenic transformation and treatment with an EZH2 inhibitor.
  • Methods of the disclosure may be used to treat a subject who has one or more mutations in a HAT that decrease/inhibit the ability of the HAT to acetylate one or more lysines of histone 3 (H3) (e.g., H3K27) or who has one or more cells with one or more mutations in a HAT that decrease/inhibit the ability of the HAT to acetylate one or more lysines of histone 3 (H3) (e.g., H3K27).
  • HAT expression and/or HAT function may be evaluated by fluorescent and non- fluorescent immunohistochemistry (IHC) methods, including well known to one of ordinary skill in the art.
  • the method comprises: (a) obtaining a biological sample from the subject; (b) contacting the biological sample or a portion thereof with an antibody that specifically binds HAT; and (c) detecting an amount of the antibody that is bound to HAT.
  • HAT expression and/or HAT function may be evaluated by a method comprising: (a) obtaining a biological sample from the subject; (b) sequencing at least one DNA sequence encoding a HAT protein from the biological sample or a portion thereof; and (c) determining if the at least one DNA sequence encoding a HAT protein contains a mutation affecting the expression and/or function of the HAT protein.
  • HAT expression or a function of HAT may be evaluated by detecting an amount of the antibody that is bound to HAT and by sequencing at least one DNA sequence encoding a HAT protein, optionally, using the same biological sample from the subject.
  • Example 1 Molecular Predictors of Response to EZH2 inhibition in NHL patients
  • EZH2 enhancer of zeste homolog 2
  • a hi stone methyl transferase enhancer of zeste homolog 2
  • Tazemetostat is an oral, first-in-class, potent, and highly selective inhibitor of EZH2 currently in phase 2 clinical development for the treatment of relapsed or refractory non- Hodgkin lymphoma (NHL).
  • NCT01897571 An ongoing phase 2 study (NCT01897571) is enrolling patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) or follicular lymphoma (FL) to assess the efficacy and safety of tazemetostat. Patients with tumors exhibiting mutated or wild-type EZH2 are eligible to participate. The primary study endpoint is objective overall response rate.
  • DLBCL diffuse large B-cell lymphoma
  • FL follicular lymphoma
  • a molecular analysis of tumor material obtained in the phase 2 study and its associations with preliminary response data is described herein, including, inter alia, a molecular predictor and a molecular predictor profile for responsiveness of a tumor or subject to treatment with an EZH2 inhibitor, e.g., with tazemetostat.
  • NGS Next generation sequencing
  • MYD88 Activating mutations in EZH2 and MYD88 (e.g., MYD88 L273P) were mutually exclusive. Accordingly, MYD88 was identified as a biomarker for response to treatment with EZH2 inhibitors, e.g., with tazemetostat, in wild-type EZH2 patients.
  • the molecular genetic profiling of NHL patients described here identifies predictors and predictor profiles for the response to treatment with EZH2 inhibitors, e.g., with tazemetostat.
  • EZH2 inhibitors e.g., with tazemetostat.
  • results disclosed herein demonstrate that plasma-based ctDNA screening is a viable method to identify NHL patients with mutations, e.g., with EZH2-activating mutations, in the absence of archive tumor samples

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Health & Medical Sciences (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The disclosure provides a method of treating cancer in a subject in need thereof including administering to the subject a therapeutically-effective amount of an enhancer of a zeste homolog 2 (EZH2) inhibitor. In certain embodiments of this method, the subject has one or more mutations, or exhibits a genetic profile, listed in Tables 1-5, and/or Figures 1-3.

Description

INHIBITORS OF EZH2 AND METHODS OF USE THEREOF
RELATED APPLICATION
[001] This application claims priority to, and the benefit of, U.S. Provisional Application No. 62/519,078, filed June 13, 2017, the contents of each of which are incorporated herein by reference in their entireties.
SEQUENCE LISTING
[002] The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled "EPIZ-083001WO_ST25.txt", which is 277,968 bytes in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.
BACKGROUND
[01] There is a long-felt yet unmet need for effective treatments for certain cancers caused by genetic alterations that result in EZH2-dependent oncogenesis.
SUMMARY
[02] In some aspects, the disclosure provides a method of treating cancer comprising administering a therapeutically effective amount of an inhibitor of Enhancer to Zeste Homolog 2 (EZH2) to a subject in need thereof, wherein the cancer is characterized by at least one mutation in one or more sequences encoding a gene or gene product listed in Tables 1 -5, and/or Figures 1- 3. In some embodiments, the cancer is characterized by at least one mutation in one or more sequences encoding: EZH2, MYD88, STAT6A, MYC, TP53, and/or HIST1H1E. In some embodiments, the cancer is characterized by a genotype that satisfies Genetic Profile 1 of Figure 3. In some embodiments, the cancer is characterized by a genotype that satisfies Genetic Profile 2 of Table 5. In some embodiments, the cancer comprises a MYD88 gain-of-function mutation. In some embodiments, the cancer does not comprise an EZH2 gain-of-function mutation. In some embodiments, the at least one mutation decreases the function of a protein encoded by the mutated sequence as compared to the function of the protein encoded by the wild-type sequence. In some embodiments, the at least one mutation is a loss-of-function mutation. In some embodiments, the method further comprises detecting the at least one mutation in the subject. In some embodiments, the detecting comprises subjecting a sample obtained from the subject to a sequence analysis assay. In some embodiments, the analysis assay is a cobas® assay. In some embodiments, the detecting comprises obtaining a sample from the subject. In some
embodiments, the sample is a tumor sample or a sample that comprises a cancer cell. In some embodiments, the sample is a sample comprising ctDNA. In some embodiments, the inhibitor of
EZH2 is
thereof.
Figure imgf000004_0001
[03] In some embodiments, the inhibitor of EZH2 is administered orally. In some
embodiments, the inhibitor of EZH2 is formulated as a tablet. In some embodiments, the therapeutically effective amount of the inhibitor of EZH2 is between 100 mg and 3200 mg per day. In some embodiments, the therapeutically effective amount of the inhibitor of EZH2 is 100 mg, 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, 1400 mg, 1600 mg or 3200 mg per day. In some embodiments, the therapeutically effective amount is 1600 mg per day. In some embodiments, the cancer expresses a wild type EZH2 protein and does not express a mutant
EZH2 protein. In some embodiments, the cancer expresses a mutant EZH2 protein. In some embodiments, the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1. In some embodiments, the mutant
EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 682 of SEQ ID NO: 1. In some embodiments, the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 692 of SEQ ID
NO: 1. In some embodiments, the at least one mutation comprises a MYD88 and/or STAT6A mutation, preferably wherein the MYD88 mutation is a gain-of-function mutation, preferably a
MYD88 L273 mutation. In some embodiments, the cancer does not have a MYC and/or a
HIST1H1E mutation. In some embodiments, the cancer (a) has a MYD88 gain-of-function or a
STAT6A loss-of-function mutation, and (b) has neither a MYC nor a HIST1H1E mutation. In some embodiments, the cancer (a) has a MYD88 L273 mutation or a STAT6A loss-of-function mutation, (b) has neither a MYC nor a HIST1H1E mutation, and (c) does not have an EZH2 gain-of-function mutation. In some embodiments, the subject is a human subject. In some embodiments, the subject has cancer. In some embodiments, the cancer is B-cell lymphoma. In some embodiments, the B-cell lymphoma is an activated B-cell (ABC) type. In some
embodiments, the B-cell lymphoma is a germinal B-cell (GBC) type. In some embodiments, the cancer is follicular lymphoma.
[04] Some aspects of this disclosure provide methods comprising selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of at least one mutation associated with a positive response to such treatment in the subject and/or based on the absence of at least one mutation associated with no response or with a negative response to such treatment in the subject. In some embodiments, the at least one mutation associated with a positive response comprise (a) an EZH2 gain-of-function mutation or a MYD88 gain-of-function mutation; and (b) a STAT6 loss-of-function mutation. In some embodiments, the at least one mutation associated with no response or with a negative response comprise (a) a MYC mutation; (b) a TP53 mutation; and/or (c) a HIST1H1E mutation. In some embodiments, the method comprises detecting the at least one mutation associated with a positive response and/or the at least one mutation associated with no response or a negative response in a sample obtained from the subject. In some embodiments, the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having either an EZH2 or a MYD88 gain-of-function mutation, or a STAT6A loss-of-function mutation; and (b) having neither a MYC mutation, nor a TP53 mutation, nor a HIST1H1E mutation. In some embodiments, the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having a MYD88 L273 mutation or a STAT6A mutation, and (b) having neither a MYC mutation, nor a TP53 mutation, nor a HIST1H1E mutation. In some embodiments, the at least one mutation consists of a single mutation. In some embodiments, the at least one mutation comprises 2 mutations or more. In some embodiments, the at least one mutation comprises 3 mutations or more. In some embodiments, the at least one mutation comprises 4 mutations or more.
[05] Some aspects of this disclosure provide methods comprising selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of a mutation profile in the subject that comprises one or more mutations described for a patient exhibiting a complete or partial response or stable disease in any of Tables 1-5 or Figures 1-3, or that comprises, encompasses, or matches the complete mutation profile of such a patient, or that comprises, encompasses, or matches Genetic Profile 1 or Genetic Profile 2 of Table 5. .
[06] The methods disclosed herein may have one or more of the following features.
[07] Exemplary sequences associated with the genes and proteins provided herein include, without limitation: MYD88 (e.g., GenBank Accession No. NM OOl 172567.1, NM 002468.4, NM_001172568.1, NM_001172569.1, and NM_001172566.1), STAT6A (e.g., GenBank Accession No. NM OO 1178078.1, NM 003153.4, NM_001178079.1, NM OO 1178080.1, or NM_001178081.1), SOCS1 (e.g., GenBank Accession No. NM_003745.1), MYC (e.g., GenBank Accession No. NM 002467.4), fflSTlHlE (e.g., GenBank Accession No.
NM_005321.2), ABLl (e.g., GenBank Accession No. NM_005157), ACVR1 (e.g., GenBank Accession No. NM_001105.4), AKTl (e.g., GenBank Accession No. NM_001014431.1), AKT2 (e.g., GenBank Accession No. NM 001243027.2), ALK (e.g., GenBank Accession No.
NM_004304.4), APC (e.g., GenBank Accession No. NM_000038.5), AR (e.g., GenBank Accession No. NM 000044.3), ARID1A (e.g., GenBank Accession No. NM 006015.4), ARID IB (e.g., GenBank Accession No. NM 020732.3), ASXL1 (e.g., GenBank Accession No. NM_015338.5), ATM (e.g., GenBank Accession No. NM_000051.3), ATRX (e.g., GenBank Accession No. NM_000489.4), AURKA (e.g., GenBank Accession No. NM_003600.3), ΑΧΓΝ2 (e.g., GenBank Accession No. NM 004655.3), BAPl (e.g., GenBank Accession No.
NM_004656.3), BCL2 (e.g., GenBank Accession No. NM_000633.2), BCR (e.g., GenBank Accession No. X02596.1), BLM (e.g., GenBank Accession No. NM_000057.3), BMPR1A (e.g., GenBank Accession No. NM 004329.2), BRAF (e.g., GenBank Accession No. NM 004333.4), BRCA1 (e.g., GenBank Accession No. NM_007294.3), BRCA2 (e.g., GenBank Accession No. NM_000059.3), BRIPl (e.g., GenBank Accession No. NM_032043.21), BTK (e.g., GenBank Accession No. NM_001287344.1), BUB IB (e.g., GenBank Accession No. NM_001211.5), CALR (e.g., GenBank Accession No. NM 004343.3), CBL (e.g., GenBank Accession No. NM_005188.3), CCND1 (e.g., GenBank Accession No. NM_053056.2), CCNE1 (e.g., GenBank Accession No. NM_001322262.1), CDC73 (e.g., GenBank Accession No. NM_024529.4), CDH1 (Accession No. NM_001317186.1), CDK4 (e.g., GenBank Accession No.
NM_000075.3), CDK6 (e.g., GenBank Accession No. NM OOl 145306.1), CDKN1B (e.g., GenBank Accession No. NM 004064.4), CDKN2A (e.g., GenBank Accession No.
NM_001195132.1), CDKN2B (e.g., GenBank Accession No. NM 078487.2), CDKN2C (e.g., GenBank Accession No. NM 078626.2), CEBPA (e.g., GenBank Accession No. NM_001285829.1), CHEK2 (e.g., GenBank Accession No. NM_145862.2), CIC (e.g., GenBank Accession No. NM_015125.4), CREBBP (e.g., GenBank Accession No. NM_001079846.1), CSF1R (e.g., GenBank Accession No. NM 001288705.2), CTNNB1 (e.g., GenBank Accession No. NM_001098209.1), CYLD (e.g., GenBank Accession No. NM 001042355.1), DAXX (Accession No. NM 001141969.1), DDB2 (e.g., GenBank Accession No. NM 001300734.1), DDR2 (e.g., GenBank Accession No. NM 001014796.1), DICERl (e.g., GenBank Accession No. NM_001291628.1), DNMT3A (e.g., GenBank Accession No. NM 001320893.1), EGFR (e.g., GenBank Accession No. NM 001346900.1), EP300 (e.g., GenBank Accession No.
NM_001429.3), ERBB2 (e.g., GenBank Accession No. NM_001289936.1), ERBB3 (e.g., GenBank Accession No. NM OO 1982.3), ERBB4 (e.g., GenBank Accession No.
NM_005235.2), ERCC1 (e.g., GenBank Accession No. NM 001166049.1), ERCC2 (e.g., GenBank Accession No. NM 001130867.1), ERCC3 (e.g., GenBank Accession No.
NM_001303418.1), ERCC4 (Accession No. NM_005236.2), ERCC5 (e.g., GenBank Accession No. NM_000123.3), ESR1 (e.g., GenBank Accession No. NM_001291241.1), ETV1 (e.g., GenBank Accession No. NM_001163147.1), ETV5 (Accession No. NM_004454.2), EWSR1 (e.g., GenBank Accession No. NM 001163287.1), EXT1 (e.g., GenBank Accession No.
NM_000127.2), EXT2 (Accession No. NM_001178083.1), FANCA (e.g., GenBank Accession No. NM_001286167.1), FANCB (Accession No. NM 001324162.1), FANCC (e.g., GenBank Accession No. NM_001243744.1), FANCD2 (e.g., GenBank Accession No. NM_001319984.1), FANCE (e.g., GenBank Accession No. NM_021922.2), FANCF (e.g., GenBank Accession No NM 022725.3.), FANCG (e.g., GenBank Accession No. NM_004629.1), FANCI (e.g.,
GenBank Accession No. NM_018193.2), FANCL (Accession No. NM_001114636.1), FANCM (e.g., GenBank Accession No. NM 001308133.1), FBXW7 (e.g., GenBank Accession No. NM_018315.4), FGFR1 (Accession No.) NM_001174065.1, FGFR2 (e.g., GenBank Accession No. NM_000141.4), FGFR3 (e.g., GenBank Accession No. NM_001163213.1), FGFR4 (e.g., GenBank Accession No. NM 213647.2), FH (e.g., GenBank Accession No. NM 000143.3), FLCN (e.g., GenBank Accession No. NM 144606.5), FLT3 (e.g., GenBank Accession No. NM_004119.2), FLT4 (e.g., GenBank Accession No. NM_002020.4), FOXL2 (e.g., GenBank Accession No. NM_023067.3), GATA1 (e.g., GenBank No. NM_002049.3), GATA2 (e.g., GenBank Accession No. NM OOl 145662.1), GNA11 (e.g., GenBank Accession No.
NM_002067.4), GNAQ (e.g., GenBank Accession No. NM_002072.4), GNAS (e.g., GenBank Accession No. NM_080425.3), GPC3 (e.g., GenBank Accession No. NM_001164619.1), H3F3A (e.g., GenBank Accession No. NM 002107.4), H3F3B (e.g., GenBank Accession No. NM_005324.4), HNF1A (e.g., GenBank Accession No. NM_000545.6), HRAS (e.g., GenBank Accession No. NM_001130442.2), IDH1 (e.g., GenBank Accession No. NM_001282387.1), IDH2 (e.g., GenBankAccessionNo. NM_001290114.1), IGF1R (e.g., GenBank Accession No. NM_001291858.1), IGF2R (e.g., GenBank Accession No. NM 000876.3), KZFl (e.g., GenBank AccessionNo. NM 001291847.1), JAK1 (e.g., GenBank Accession No.
NM 001321857.1), JAK2 (e.g., GenBank AccessionNo. NM_001322195.1), JAK3 (e.g., GenBank Accession No. NM 000215.3), KDR (e.g., GenBank Accession No. NM 002253.2), KIT (e.g., GenBank Accession No. NM 001093772.1), KRAS (e.g., GenBank Accession No. NM_033360.3), MAML1 (e.g., GenBank Accession No. NM_014757.4), MAP2K1 (e.g., GenBank Accession No. NM 002755.3), MAP2K4 (e.g., GenBank Accession No.
NM 001281435.1), MDM2 (e.g., GenBank Accession No. NM_001145337.2), MDM4 (e.g., GenBank Accession No. NM 001278519.1), MED12 (e.g., GenBank Accession No.
NM_005120.2), MEN1 (e.g., GenBank Accession No. NM_130804.2), MET (e.g., GenBank Accession No NM 000245.3), MLH1 (e.g., GenBank Accession No. NM 000249.3), MLL (e.g., GenBank AccessionNo. AF232001.1), MPL (e.g., GenBank AccessionNo.
NM_005373.2), MSH2 (e.g., GenBank Accession No. NM_000251.2), MSH6 (e.g., GenBank Accession No. NM_000179.2), MTOR (Accession No. NM_004958.3), MUTYH (e.g., GenBank Accession No. NM 001048171.1), MYC (e.g., GenBank Accession No.
NM_002467.4), MYCL1 (e.g., GenBank Accession No NM 001033081.2), MYCN (e.g., GenBank Accession No. NM 001293231.1), NBN (e.g., GenBank Accession No.
NM_001024688.2), NCOA3 (e.g., GenBank Accession No. NM OOl 174087.1), NFl (e.g., GenBank Accession No. NM OO 1042492.2), NF2 (e.g., GenBank Accession No.
NM_181831.2), NKX2-l(e.g., GenBank Accession No. NM_001079668.2), NOTCH1 (e.g., GenBank Accession No. NM 017617.4), NOTCH2 (e.g., GenBank Accession No
NM_001200001.1), NOTCH3 (e.g., GenBank Accession No. NM_000435.2), NOTCH4 (Accession No. NR_134950.1), NPM1 (e.g., GenBank Accession No. NM_002520.6), NRAS (Accession No. NM_002524.4), NTRKl (e.g., GenBank AccessionNo. NM_001007792.1), PALB2 (e.g., GenBank Accession No. NM 024675.3), PAX5 (e.g., GenBank Accession No. NM 001280552.1), PBRMl (e.g., GenBank AccessionNo. NM_181042.4), PDGFRA (e.g., GenBank Accession No. NM 006206.4), PHOX2B (e.g., GenBank Accession No.
NM_003924.3), PIK3CA (e.g., GenBank Accession No. NM_006218.3), PIK3R1 (Accession No. NM_001242466.1), PMS1 (e.g., GenBank Accession No. NM_001321051.1), PMS2 (e.g., GenBank Accession No. NM 000535.6), POLD1 (e.g., GenBank Accession No.
NM 001308632.1), POLE (e.g., GenBank Accession No. NM_006231.3), POLH (e.g., GenBank Accession No. NM_001291970.1), POT1 (e.g., GenBank Accession No. NM_001042594.1), PRKAR1A (e.g., GenBank Accession No. NM 001278433.1), PRSS1 (e.g., GenBank Accession No. NM_002769.4), PTCH1 (e.g., GenBank Accession No. NM_000264.3), PTEN (e.g., GenBank Accession No. NM 000314.6), PTPN11 (e.g., GenBank Accession No.
NM_001330437.1), RAD51C (e.g., GenBank Accession No. NR_103873.1), RAF1 (e.g., GenBank Accession No. NM 002880.3), RBI (e.g., GenBank Accession No. NM 000321.2), RECQL4 (e.g., GenBank Accession No. NM 004260.3), RET (e.g., GenBank Accession No.), RNF43(e.g., GenBank Accession No. NM_017763.5), ROS1 (e.g., GenBank Accession No. NM 002944.2), RUNX1 (e.g., GenBank Accession No. NM_001122607.1), SBDS (e.g., GenBank Accession No. NM 016038.2), SDHAF2 (e.g., GenBank Accession No.
NM_017841.2), SDHB (e.g., GenBank Accession No.), SDHC (e.g., GenBank Accession No.), SDHD (e.g., GenBank Accession No. NM 001276503.1), SF3B1 (e.g., GenBank Accession No. NM 001308824.1), SMAD2 (e.g., GenBank Accession No. NM_001135937.2), SMAD3 (e.g., GenBank Accession No. NM 001145104.1), SMAD4 (e.g., GenBank Accession No.
NM_005359.5), SMARCB1 (e.g., GenBank Accession No. NM_001007468.2), SMO (e.g., GenBank Accession No. NM 005631.4), SRC (e.g., GenBank Accession No. NM 005417.4), STAG2 (e.g., GenBank Accession No. NM_001282418.1), STKll (e.g., GenBank Accession No. NM_000455.4), SUFU (e.g., GenBank Accession No. NM_001178133.1), TERT (e.g., GenBank Accession No. NM_001193376.1), TET2 (e.g., GenBank Accession No.
NM_017628.4), TGFBR2 (e.g., GenBank Accession No. NM_001024847.2), TNFAIP3 (e.g., GenBank Accession No. NM OO 1270508.1), TOPI (e.g., GenBank Accession No.
NM_003286.3), TP53 (e.g., GenBank Accession No. NM_000546.5), TSC1 (e.g., GenBank Accession No. NM_001162427.1), TSC2 (e.g., GenBank Accession No. NM_001318832.1), TSHR (e.g., GenBank Accession No. NM_000369.2), VHL (e.g., GenBank Accession No. NM_000551.3), WAS (e.g., GenBank Accession No. NM 000377.2), WRN (e.g., GenBank Accession No. NM_000553.4), WT1 (e.g., GenBank Accession No. NM_000378.4), XPA (e.g., GenBank Accession No. NM 000380.3), XPC (e.g., GenBank Accession No. NM 004628.4), and/or XRCC1 (e.g., GenBank Accession No. NM 006297.2). It will be understood that the sequences provided above and elsewhere herein are exemplary, and serve to illustrate sequences suitable for some embodiments of the present disclosure. It will also be understood that, in some embodiments, the sequence encoding the gene product referred to herein is a genomic DNA sequence. The skilled artisan will be aware of additional suitable sequences beyond the exemplary, non-limiting sequences provided above, for each gene or gene product (e.g., transcript, mRNA, or protein) referred to herein, or will be able to ascertain such suitable sequences without more than routine effort based on the present disclosure and the knowledge in the art.
[08] In some embodiments, the at least one mutation decreases the function of a protein encoded by the mutated sequence as compared to the function of the protein encoded by the wild-type sequence. In some embodiments, the at least one mutation is a loss-of-function mutation.
[09] In some embodiments, the method further comprises detecting the at least one mutation in the subject.
[010] In some embodiments, the detecting comprises subjecting a sample obtained from the subject to a sequence analysis assay.
[011] In some embodiments, the inhibitor of EZH2 is
Figure imgf000010_0001
or a pharmaceutically-acceptable salt thereof.
[012] In some embodiments, the inhibitor of EZH2 is administered orally.
[013] In some embodiments, the inhibitor of EZH2 is formulated as a tablet.
[014] In some embodiments, the therapeutically effective amount of the inhibitor of EZH2 is between 100 mg and 3200 mg per day. -In some embodiments, the therapeutically effective amount of the inhibitor of EZH2 is 100 mg, 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, 1400 mg, 1600 mg or 3200 mg per day. In some embodiments, the therapeutically effective amount is 1600 mg per day. In some embodiments, the therapeutically effective amount of the inhibitor of is administered at 800 mg twice per day (BID).
[015] In some embodiments, the subject or cancer expresses a wild type EZH2 protein and/or does not express a mutant EZH2 protein.
[016] In some embodiments, the subject or cancer expresses a mutant EZH2 protein. In some embodiments, the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1. In some embodiments, the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 682 of SEQ ID NO: 1. In some embodiments, the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 692 of SEQ ID NO: 1.
[017] In some embodiments, the at least one mutation consists of a single mutation. In some embodiments, the at least one mutation comprises 2 mutations or more. In some embodiments, the at least one mutation comprises 3 mutations or more. In some embodiments, the at least one mutation comprises 4 mutations or more. In some embodiments, the at least one mutation comprises S mutations or more.
[018] In some embodiments, the at least one mutation comprises 2 mutations, 3 mutations, 4 mutations, 5 mutations, 6 mutations, 7 mutations, 8 mutations, 9 mutations, 10 mutations, 11 mutations, 12 mutations, 13 mutations, 14 mutations, IS mutations, 16 mutations, 17 mutations, 18 mutations, 19 mutations, or 20 mutations.
[019] In some embodiments, the at least one mutation comprises at least one positive mutation (e.g., with or without a negative mutation). In some embodiments, the at least one mutation comprises at least one negative mutation (e.g., with or without a positive mutation). In some embodiments, the at least one mutation comprises both positive and negative mutations. The term "positive mutation", as used herein, refers to a mutation that sensitizes a subject, a cancer, or malignant cell or population of cells, to EZH2 treatment, or, in some embodiments, that renders a subject, cancer, or malignant cell or population of cells, more sensitive to EZH2 treatment. The term "negative mutation", as used herein, refers to a mutation that desensitizes a subject, a cancer, or malignant cell or population of cells, to EZH2 treatment, or, in some embodiments, that renders a subject, cancer, or malignant cell or population of cells, less sensitive to EZH2 treatment. In some embodiments, the disclosure provides a method of identifying molecular variants in tumor samples harvested from NHL patients treated with a compound of the disclosure. In some embodiments, the disclosure provides a method of identifying molecular variants in cell free circulating tumor DNA (ctDNA) harvested from NHL patients treated with a compound of the disclosure.
[020] In some embodiments, the molecular variants identified therein may correlate with clinical response, minimal residual disease or emergence of resistance.
[021] The summary above is meant to illustrate, in a non-limiting manner, some of the embodiments, advantages, features, and uses of the technology disclosed herein. Other embodiments, advantages, features, and uses of the technology disclosed herein will be apparent from the Detailed Description, the Drawings, the Examples, and the Claims.
BRIEF DESCRIPTION OF THE DRAWINGS
[022] The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
[023] The above and further features will be more clearly appreciated from the following detailed description when taken in conjunction with the accompanying drawings.
[024] Figure 1 shows an overview of the analytical and statistical methods used in this study.
[025] Figures 2A-2D show somatic mutations, amplifications, and translocations detected in 92 NHL patients by best overall response. Figure 2A shows all 92 NHL patients assessed. Figure 2B shows NHL patients classified as responders (complete response (CR) or partial response (PR)). Figure 2C shows FL patients that were classified as non-responders. Figure 2D shows DLBCL patients that were classified as non-responders. NHL: Non-Hodgkin Lymphoma; CR: complete response; PR: partial response; SD: stable disease; PD: progressive disease; UK: unknown response. Patients with CR and PR were classified as responders. Patients with SD, PD, and UK were classified as non-responders.
[026] Figure 3 shows variants identified as significantly associated with tazemetostat response using gene level analysis of archive tumor tissue distributed by response-favoring mutant and response favoring wild-type gene (N=92).
[027] Figure 4 is a structure model of partial EZH2 protein based on the A chain of nuclear receptor binding SET domain protein 1 (NSD1). This model corresponds to amino acid residues 533-732 of EZH2 sequence of SEQ ID NO: 1. DETAILED DESCRIPTION
[028] Tazemetostat demonstrates clinical activity as a monotherapy in patients with relapsed or refractory DLBCL (both GCB and non-GCB), follicular lymphoma (FL) and marginal zone lymphomas (MZL). Objective responses in tumors with either wild-type or mutation in EZH2 are durable as patients are ongoing at 7+ to 21+ months. Safety profile as monotherapy continues to be acceptable and favorable for combination development. Recommended phase II dose (RP2D) of 800 mg BID supported by safety, efficacy, PK and PD.
[029] Baseline somatic mutation profiling revealed associations between objective response to tazemetostat and genetic alterations, e.g., mutations in genomic sequences encoding MYD88, STAT6A, SOCS1, MYC, TP53, fflSTlHlE, and histone acetyltransferases, such as, for example CREBBP and EP300.
EZH2
[030] EZH2 is a histone methyltransferase that is the catalytic subunit of the PRC2 complex which catalyzes the mono- through tri-methylation of lysine 27 on histone H3 (H3-K27).
[031] Point mutations of the EZH2 gene at a single amino acid residue (e.g., Tyr641, herein referred to as Y641) of EZH2 have been reported to be linked to subsets of human B-cell lymphoma. Morin et al. (2010) Nat Genet 42(2): 181-5. In particular, Morin et al. reported that somatic mutations of tyrosine 641 (Y641F, Y641H, Y641N, and Y641S) of EZH2 were associated with follicular lymphoma (FL) and the germinal center B cell-like (GCB) subtype of diffuse large B-cell lymphoma (DLBCL). The mutant allele is always found associated with a wild-type allele (heterozygous) in disease cells, and the mutations were reported to ablate the enzymatic activity of the PRC2 complex for methylating an unmodified peptide substrate.
[032] The mutant EZH2 refers to a mutant EZH2 polypeptide or a nucleic acid sequence encoding a mutant EZH2 polypeptide. Preferably the mutant EZH2 comprises one or more mutations in its substrate pocket domain as defined in SEQ ID NO: 6. For example, the mutation may be a substitution, a point mutation, a nonsense mutation, a missense mutation, a deletion, or an insertion. Exemplary substitution amino acid mutation includes a substitution at amino acid position 677, 687, 674, 685, or 641 of SEQ ID NO: 1, such as, but is not limited to a substitution of glycine (G) for the wild type residue alanine (A) at amino acid position 677 of SEQ ID NO: 1 (A677G); a substitution of valine (V) for the wild type residue alanine (A) at amino acid position 687 of SEQ ID NO: 1 (A687V); a substitution of methionine (M) for the wild type residue valine (V) at amino acid position 674 of SEQ ID NO: 1 (V674M); a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 685 of SEQ ID NO: 1 (R685H); a substitution of cysteine (C) for the wild type residue arginine (R) at amino acid position 685 of SEQ ID NO: 1 (R685C); a substitution of phenylalanine (F) for the wild type residue tyrosine (Y) at amino acid position 641 of SEQ ID NO: 1 (Y641F); a substitution of histidine (H) for the wild type residue tyrosine (Y) at amino acid position 641 of SEQ ID NO: 1 (Y641H); a substitution of asparagine (N) for the wild type residue tyrosine (Y) at amino acid position 641 of SEQ ID NO: 1 (Y641 N); a substitution of serine (S) for the wild type residue tyrosine (Y) at amino acid position 641 of SEQ ID NO: 1 (Y641S); or a
substitution of cysteine (C) for the wild type residue tyrosine (Y) at amino acid position 641 of SEQ ID NO: 1 (Y641C).
[033] The mutation may also include a substitution of serine (S) for the wild type residue asparagine (N) at amino acid position 322 of SEQ ID NO: 3 (N322S), a substitution of glutamine (Q) for the wild type residue arginine (R) at amino acid position 288 of SEQ ID NO: 3 (R288Q), a substitution of isoleucine (I) for the wild type residue threonine (T) at amino acid position 573 of SEQ ID NO: 3 (T573I), a substitution of glutamic acid (E) for the wild type residue aspartic acid (D) at amino acid position 664 of SEQ ID NO: 3 (D664E), a substitution of glutamine (Q) for the wild type residue arginine (R) at amino acid position 458 of SEQ ID NO: 5 (R458Q), a substitution of lysine (K) for the wild type residue glutamic acid (E) at amino acid position 249 of SEQ ID NO: 3 (E249K), a substitution of cysteine (C) for the wild type residue arginine (R) at amino acid position 684 of SEQ ID NO: 3 (R684C), a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 628 of SEQ ID NO: 21 (R628H), a substitution of histidine (H) for the wild type residue glutamine (Q) at amino acid position 501 of SEQ ID NO: 5 (Q501 H), a substitution of asparagine (N) for the wild type residue aspartic acid (D) at amino acid position 192 of SEQ ID NO: 3 (D192N), a substitution of valine (V) for the wild type residue aspartic acid (D) at amino acid position 664 of SEQ ID NO: 3 (D664V), a substitution of leucine (L) for the wild type residue valine (V) at amino acid position 704 of SEQ ID NO: 3 (V704L), a substitution of serine (S) for the wild type residue proline (P) at amino acid position 132 of SEQ ID NO: 3 (P132S), a substitution of lysine (K) for the wild type residue glutamic acid (E) at amino acid position 669 of SEQ ID NO: 21 (E669K), a substitution of threonine (T) for the wild type residue alanine (A) at amino acid position 255 of SEQ ID NO: 3 (A255T), a substitution of valine (V) for the wild type residue glutamic acid (E) at amino acid position 726 of SEQ ID NO: 3 (E726V), a substitution of tyrosine (Y) for the wild type residue cysteine (C) at amino acid position 571 of SEQ ID NO: 3 (C571 Y), a substitution of cysteine (C) for the wild type residue phenylalanine (F) at amino acid position 145 of SEQ ID NO: 3 (F145C), a substitution of threonine (T) for the wild type residue asparagine (N) at amino acid position 693 of SEQ ID NO: 3 (N693T), a substitution of serine (S) for the wild type residue phenylalanine (F) at amino acid position 145 of SEQ ID NO: 3 (F145S), a substitution of histidine (H) for the wild type residue glutamine (Q) at amino acid position 109 of SEQ ID NO: 21 (Q109H), a substitution of cysteine (C) for the wild type residue phenylalanine (F) at amino acid position 622 of SEQ ID NO: 21 (F622C), a substitution of arginine (R) for the wild type residue glycine (G) at amino acid position 135 of SEQ ID NO: 3 (G135R), a substitution of glutamine (Q) for the wild type residue arginine (R) at amino acid position 168 of SEQ ID NO: 5 (R168Q), a substitution of arginine (R) for the wild type residue glycine (G) at amino acid position 159 of SEQ ID NO: 3 (G159R), a substitution of cysteine (C) for the wild type residue arginine (R) at amino acid position 310 of SEQ ID NO: 5 (R3 IOC), a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 561 of SEQ ID NO: 3 (R561H), a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 634 of SEQ ID NO: 21 (R634H), a substitution of arginine (R) for the wild type residue glycine (G) at amino acid position 660 of SEQ ID NO: 3 (G660R), a substitution of cysteine (C) for the wild type residue tyrosine (Y) at amino acid position 181 of SEQ ID NO: 3 (Y181C), a substitution of arginine (R) for the wild type residue histidine (H) at amino acid position 297 of SEQ ID NO: 3 (H297R), a substitution of serine (S) for the wild type residue cysteine (C) at amino acid position 612 of SEQ ID NO: 21 (C612S), a substitution of tyrosine (Y) for the wild type residue histidine (H) at amino acid position 694 of SEQ ID NO: 3
(H694Y), a substitution of alanine (A) for the wild type residue aspartic acid (D) at amino acid position 664 of SEQ ID NO: 3 (D664A), a substitution of threonine (T) for the wild type residue isoleucine (I) at amino acid position 150 of SEQ ID NO: 3 (I150T), a substitution of arginine (R) for the wild type residue isoleucine (I) at amino acid position 264 of SEQ ID NO: 3 (I264R), a substitution of leucine (L) for the wild type residue proline (P) at amino acid position 636 of SEQ ID NO: 3 (P636L), a substitution of threonine (T) for the wild type residue isoleucine (I) at amino acid position 713 of SEQ ID NO: 3 (I713T), a substitution of proline (P) for the wild type residue glutamine (Q) at amino acid position 501 of SEQ ID NO: 5 (Q501P), a substitution of glutamine (Q) for the wild type residue lysine (K) at amino acid position 243 of SEQ ID NO: 3 (K243Q), a substitution of aspartic acid (D) for the wild type residue glutamic acid (E) at amino acid position 130 of SEQ ID NO: 5 (E130D), a substitution of glycine (G) for the wild type residue arginine (R) at amino acid position 509 of SEQ ID NO: 3 (R509G), a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 566 of SEQ ID NO: 3 (R566H), a substitution of histidine (H) for the wild type residue aspartic acid (D) at amino acid position 677 of SEQ ID NO: 3 (D677H), a substitution of asparagine (N) for the wild type residue lysine (K) at amino acid position 466 of SEQ ID NO: 5 (K466N), a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 78 of SEQ ID NO: 3 (R78H), a substitution of methionine (M) for the wild type residue lysine (K) at amino acid position 1 of SEQ ID NO: 6 (K6M), a substitution of leucine (L) for the wild type residue serine (S) at amino acid position 538 of SEQ ID NO: 3 (S538L), a substitution of glutamine (Q) for the wild type residue leucine (L) at amino acid position 149 of SEQ ID NO: 3 (L149Q), a substitution of valine (V) for the wild type residue leucine (L) at amino acid position 252 of SEQ ID NO: 3 (L252V), a substitution of valine (V) for the wild type residue leucine (L) at amino acid position 674 of SEQ ID NO: 3 (L674V), a substitution of valine (V) for the wild type residue alanine (A) at amino acid position 656 of SEQ ID NO: 3 (A656V), a substitution of aspartic acid (D) for the wild type residue alanine (A) at amino acid position 731 of SEQ ID NO: 3 (Y73 ID), a substitution of threonine (T) for the wild type residue alanine (A) at amino acid position 345 of SEQ ID NO: 3 (A345T), a substitution of aspartic acid (D) for the wild type residue alanine (A) at amino acid position 244 of SEQ ID NO: 3 (Y244D), a substitution of tryptophan (W) for the wild type residue cysteine (C) at amino acid position 576 of SEQ ID NO: 3 (C576W), a substitution of lysine (K) for the wild type residue asparagine (N) at amino acid position 640 of SEQ ID NO: 3 (N640K), a substitution of lysine (K) for the wild type residue asparagine (N) at amino acid position 675 of SEQ ID NO: 3 (N675K), a substitution of tyrosine (Y) for the wild type residue aspartic acid (D) at amino acid position 579 of SEQ ID NO: 21 (D579Y), a substitution of isoleucine (I) for the wild type residue asparagine (N) at amino acid position 693 of SEQ ID NO: 3 (N693I), and a substitution of lysine (K) for the wild type residue asparagine (N) at amino acid position 693 of SEQ ID NO: 3 (N693K).
[034] The mutation may be a frameshift at amino acid position 730, 391, 461, 441, 235, 254, 564, 662, 715, 405, 685, 64, 73, 656, 718, 374, 592, 505, 730, or 363 of SEQ ID NO: 3, 5 or 21 or the corresponding nucleotide position of the nucleic acid sequence encoding SEQ ID NO: 3, 5, or 21. The mutation of the EZH2 may also be an insertion of a glutamic acid (E) between amino acid positions 148 and 149 of SEQ ID NO: 3, 5 or 21. Another example of EZH2 mutation is a deletion of glutamic acid (E) and leucine (L) at amino acid positions 148 and 149 of SEQ ID NO: 3, 5 or 21. The mutant EZH2 may further comprise a nonsense mutation at amino acid position 733, 25, 317, 62, 553, 328, 58, 207, 123, 63, 137, or 60 of SEQ ID NO: 3, 5 or 21.
[035] Human EZH2 nucleic acids and polypeptides have previously been described. See, e.g., Chen et al. (1996) Genomics 38:30-7 [746 amino acids]; Swiss-Prot Accession No. Q15910 [746 amino acids]; GenBank Accession Nos. NM 004456 and NP 004447 (isoform a [751 amino acids]); and GenBank Accession Nos. NM l 52998 and NP 694543 (isoform b [707 amino acids]), each of which is incorporated herein by reference in its entirety.
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
[036] A structure model of partial EZH2 protein based on the A chain of nuclear receptor binding SET domain protein 1 (NSD1) is provided in Figure 4. This model corresponds to amino acid residues 533-732 of EZH2 sequence of SEQ ID NO: 1.
[037] The corresponding amino acid sequence of this structure model is provided below. The residues in the substrate pocket domain are underlined. The residues in the SET domain are shown italic.
Figure imgf000021_0003
[038] The catalytic site of EZH2 is believed to reside in a conserved domain of the protein known as the SET domain. The amino acid sequence of the SET domain of EZH2 is provided by the following partial sequence spanning amino acid residues 613-726 of Swiss-Prot
Accession No. Q15910 (SEQ ID NO: 1):
Figure imgf000021_0004
[039] The tyrosine (Y) residue shown underlined in SEQ ID NO: 7 is Tyr641 (Y641) in Swiss- Prot Accession No. Q15910 (SEQ ID NO: 1).
[040] The SET domain of GenBank Accession No. NP_004447 (SEQ ID NO: 3) spans amino acid residues 618-731 and is identical to SEQ ID NO:6. The tyrosine residue corresponding to Y641 in Swiss-Prot Accession No. Q15910 shown underlined in SEQ ID NO: 7 is Tyr646 (Y646) in GenBank Accession No. NP_004447 (SEQ ID NO: 3).
[041] The SET domain of GenBank Accession No. NP_694543 (SEQ ID NO: 5) spans amino acid residues 574-687 and is identical to SEQ ID NO: 7. The tyrosine residue corresponding to
Y641 in Swiss-Prot Accession No. Q15910 shown underlined in SEQ ID NO: 7 is Tyr602
(Y602) in GenBank Accession No. NP_694543 (SEQ ID NO: 5).
[042] The nucleotide sequence encoding the SET domain of GenBank Accession No.
NP 004447 is
Figure imgf000021_0002
where the codon encoding Y641 is shown underlined.
[043] For purposes of this application, amino acid residue Y641 of human EZH2 is to be understood to refer to the tyrosine residue that is or corresponds to Y641 in Swiss-Prot
Accession No. Q 15910.
Figure imgf000021_0001
Figure imgf000022_0001
[044] A Y641 mutant of human EZH2, and, equivalently, a Y641 mutant of EZH2, is to be understood to refer to a human EZH2 in which the amino acid residue corresponding to Y641 of wild-type human EZH2 is substituted by an amino acid residue other than tyrosine.
[045] In one embodiment the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of a single amino acid residue corresponding to Y641 of wild-type human EZH2 by an amino acid residue other than tyrosine.
[046] In one embodiment the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of phenylalanine (F) for the single amino acid residue corresponding to Y641 of wild-type human EZH2. The Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641F mutant or, equivalently,
Figure imgf000022_0002
[047] In one embodiment the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of histidine (H) for the single amino acid residue corresponding to Y641 of wild-type human EZH2. The Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641H mutant or, equivalently,
Figure imgf000023_0001
[048] In one embodiment the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of asparagine (N) for the single amino acid residue corresponding to Y641 of wild-type human EZH2. The Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641 N mutant or, equivalently,
Figure imgf000023_0002
[049] In one embodiment the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of serine (S) for the single amino acid residue corresponding to Y641 of wild-type human EZH2. The Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641S mutant or, equivalently, Y641S.
Figure imgf000023_0003
Figure imgf000024_0001
[050] In one embodiment the amino acid sequence of a Y641 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of cysteine (C) for the single amino acid residue corresponding to Y641 of wild-type human EZH2. The Y641 mutant of EZH2 according to this embodiment is referred to herein as a Y641 C mutant or, equivalently, Y641C.
Figure imgf000024_0002
[051] In one embodiment the amino acid sequence of a A677 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of a non-alanine amino acid, preferably glycine (G) for the single amino acid residue corresponding to A677 of wild- type human EZH2. The A677 mutant of EZH2 according to this embodiment is referred to herein as an A677 mutant, and preferably an A677G mutant or, equivalently, A677G.
Figure imgf000024_0003
Figure imgf000025_0001
[052] In one embodiment the amino acid sequence of a A687 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of a non-alanine amino acid, preferably valine (V) for the single amino acid residue corresponding to A687 of wild-type human EZH2. The A687 mutant of EZH2 according to this embodiment is referred to herein as an A687 mutant and preferably an A687V mutant or, equivalently, A687V.
Figure imgf000025_0002
[053] In one embodiment the amino acid sequence of a R685 mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 only by substitution of a non-arginine amino acid, preferably histidine (H) or cysteine (C) for the single amino acid residue corresponding to R68S of wild-type human EZH2. The R68S mutant of EZH2 according to this embodiment is referred to herein as an R68S mutant and preferably an R68SC mutant or an R68SH mutant or, equivalently, R685H or R685C.
Figure imgf000025_0003
Figure imgf000026_0001
[054] In one embodiment the amino acid sequence of a mutant of EZH2 differs from the amino acid sequence of wild-type human EZH2 in one or more amino acid residues in its substrate pocket domain as defined in SEQ ID NO: 6. The mutant of EZH2 according to this
embodiment is referred to herein as an EZH2 mutant.
Figure imgf000026_0002
[055] Histone acetyltransferase (HAT) enzymes of the disclosure activate gene transcription by transferring an acetyl group from acetyl CoA to form ε-Ν-acetyllysine, which serves to modify hi stones and increase transcription by, for example, generating or exposing binding sites for protein-protein interaction domains.
[056] HAT enzymes of the disclosure include, but are not limited to, those enzymes of the p300/CBP family.
[057] In certain embodiments, a mutation of the disclosure may occur in a sequence encoding the p300 HAT, including the nucleotide sequence of the EP300 gene, encoding p300 (below, corresponding to GenBank Accession No. NM 001429.3, defined as Homo sapiens El A binding protein p300 (EP300), mRNA; and identified as SEQ ID NO: 19).
Figure imgf000026_0003
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
[058] In certain embodiments, a mutation of the disclosure may occur in a sequence encoding the p300 HAT, including the amino acid sequence of the p300 protein (below, corresponding to GenBank Accession No. NP 001420.2, defined as Homo sapiens El A-binding protein, 300kD; El A-associated protein p300; p300 HAT; and identified as SEQ ID NO: 20).
Figure imgf000029_0002
Figure imgf000030_0003
[059] In certain embodiments, a mutation of the disclosure may occur in a sequence encoding the CREB Binding Protein (CREBBP) HAT, including the nucleotide sequence encoding CREBBP (below, corresponding to GenBank Accession No. NM 004380, defined as Homo sapiens CREB binding protein (CREBBP), transcript variant 1, mRNA; and identified as SEQ
Figure imgf000030_0001
Figure imgf000030_0002
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
[060] In certain embodiments, a mutation of the disclosure may occur in a sequence encoding the CREB Binding Protein (CREBBP) HAT, including the amino acid sequence encoding CREBBP (below, corresponding to GenBank Accession No. NP 004371, defined as Homo sapiens CREB-binding protein isoform a; and identified as SEQ ID NO: 24).
Figure imgf000033_0002
Figure imgf000033_0003
Figure imgf000034_0001
[061] In certain embodiments, a mutation of the disclosure may occur in a sequence encoding the CREB Binding Protein (CREBBP) HAT, including the nucleotide sequence encoding CREBBP (below, corresponding to GenBank Accession No. NM OO 1079846, defined as Homo sapiens CREB binding protein (CREBBP), transcript variant 2, mRNA; and identified as SEQ ID NO: 25).
Figure imgf000034_0002
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Next Generation Sequencing
[063] The compounds of the disclosure are inhibitors of the hi stone methyltransferase EZH2 for use in the treatment of patients with non-Hodgkin lymphoma (NHL), and in patients with certain genetically defined solid tumors. Activating EZH2 mutations present in NHL patients has been implicated to predict response to EZH2 inhibition (Knutson et al., Nat. Chem. Biol. 2012; 8: 890-896, the content of which is incorporated herein by reference in its entirety). Furthermore, a phase 1 clinical trial of tazemetostat demonstrated clinical responses in both EZH2 mutant and wild type patients (ClinicalTrials.gov identifier: NCT01897571). However, the impact of somatic mutations other than EZH2 on likelihood of response to tazemetostat in NHL patients is currently unknown. In some aspects, the present disclosure provides a multi-gene NHL targeted next generation sequencing (NGS) panel (e.g., a 39-gene panel or a 62-gene panel, or a panel combining a plurality of genes or gene products referred to herein) capable of analyzing samples from malignant cells, tissues, or body fluids, e.g., archive tissue or cell-free circulating tumor DNA (ctDNA) isolated from plasma. In some aspects, the NGS panel is capable of identifying molecular variants, including specific somatic sequence mutations (single base and
insertion/deletion, e.g., EZH2), amplifications (e.g., BLC2) and translocations (e.g., BCL2 and MYC) in the tumor and ctDNA samples down to variant allele frequencies of 2% and 0.1% for archive and ctDNA respectively. For example, molecular variants associated with positive (e.g., EZH2, STAT6, MYD88, and SOCS1 mutations) and negative (e.g., MYC and HIST1H1E mutations) clinical responses to tazemetostat treatment were identified. Furthermore, sequencing of phase 1 NHL patients utilizing a 62 gene NHL NGS panel revealed a complex genetic landscape with epigenetic modifiers CREBBP and KMT2D representing the most frequently mutated genes in this sample set. Further aspects of the disclosure provide for an NGS panel with the ability to determine molecular profiles using ctDNA that enables patient
characterization where archive tumor tissue or DNA is absent or limiting. Additionally, profiling ctDNA enables longitudinal monitoring of a patient's mutation burden without the need for tumor biopsies.
[064J Without wishing to be bound by theory, mutations identified by the NGS panel disclosed herein, may be used for patient stratification. Accordingly, in some embodiments, the disclosure provides a method of selecting a patient for cancer treatment if the patient has one or more mutations disclosed herein. In some embodiments, the patient selected for the cancer treatment has two or more (e.g., two, three, four, five, six, seven, eight, or more) mutations disclosed herein.
[065] In some embodiments, a method is provided in which a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of one or more mutations associated with a positive response to such treatment in the subject, e.g., as determined by ctDNA analysis. In some embodiments, a mutation (or a combination of two or more mutations) associated with a positive response is a mutation (or a combination of mutations) that is present only in patients who responded with complete or partial response or, in some embodiments, with stable disease in any of the studies presented herein, e.g., those summarized in Figures 1-3. In some embodiments, a mutation (or a combination of two or more mutations) associated with a positive response is a mutation (or a combination of mutations) that is not randomly distributed within the patient population examined, but is overrepresented in those patients who responded with a complete or partial response or, in some embodiments, stable disease, in any of the studies presented herein, e.g., those summarized in Figures 1-3. In some embodiments, a mutation (or combination of mutations) associated with a positive response is a mutation (or combination of mutations) that is overrepresented in the responding (CR, PR, or, in some embodiments, SD) patient population at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, or at least 10-fold, as compared to the patient population that did not respond or responded with progressive disease (PD).
[066J In some embodiments, a method is provided in which a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the absence of one or more mutations associated with a negative response to such treatment in the subject, e.g., as determined by ctDNA analysis. In some embodiments, a mutation (or a combination of two or more mutations) associated with a negative response is a mutation (or a combination of mutations) that is present only in patients who did not respond or responded with progressive disease (PD) in any of the studies presented herein, e.g., those summarized in Figures 1-3. In some embodiments, a mutation (or a combination of two or more mutations) associated with a negative response is a mutation (or a combination of mutations) that is not randomly distributed within the patient population examined, but is overrepresented in those patients who did not respond or responded with progressive disease in any of the studies presented herein, e.g., those summarized in Figures 1-3. In some embodiments, a mutation (or combination of mutations) associated with a negative response is a mutation (or combination of mutations) that is overrepresented in the non-responding or progressive disease (PD) patient population at least 2-fold, at least 3-fold, at least 4-fold, at least S-fold, or at least 10-fold, as compared to the patient population that responded with CR, PR, or, in some embodiments, SD.
[067] In some embodiments, a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of two or more (e.g., two, three, four, five, six, seven, eight, or more) mutations in the subject that match the mutations observed in a profile of a patient who exhibited a complete or partial response in any of the studies described herein (e.g., those summarized in Figures 1-3). In some embodiments, a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of a mutation profile (e.g., of two or more (e.g., two, three, four, five, six, seven, eight, or more)) mutations in the subject that match the mutation profile of a patient who exhibited a complete or partial response in any of the studies described herein (e.g., those summarized in Figures 1-3). Typically, a mutation in a gene or gene product (e.g., in a transcript, mRNA, or protein) is detected by comparing a given sequence with a reference sequence, e.g., a human reference genome sequence (e.g., human reference genome hgl9), and identifying a mismatch in the sequence at hand as compared to the reference sequence.
[068] In some embodiments, a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of two or more (e.g., two, three, four, five, six, seven, eight, or more) mutations in the subject that match the mutations observed in a profile of a patient who exhibited stable disease in any of the studies described herein (e.g., those summarized in Figures 1-3). In some embodiments, a subject having cancer is selected for treatment with an EZH2 inhibitor, e.g., an EZH2 inhibitor disclosed herein, based on the presence of a mutation profile (e.g., two or more (e.g., two, three, four, five, six, seven, eight, or more)) mutations in the subject that match the mutation profile of a patient who exhibited stable disease in any of the studies described herein (e.g., those summarized in Figures 1-3).
[069] In some embodiments, methods of treating cancer is provided that comprises
administering a therapeutically effective amount of an inhibitor of EZH2 to a subject in need thereof, wherein the subject has at least one mutation in one or more sequences encoding a gene or a gene product (e.g., a transcript, mRNA, or protein) listed in Tables 1-5, and/or Figures 1-3. In some embodiments, the subject has at least one mutation in in one or more sequences encoding: MYD88, STAT6A, SOCS1, MYC, HIST1H1E, ABL1, ACVR1, AKT1, AKT2, ALK, APC, AR, ARID1A, ARID1B, ASXL1, ATM, ATRX, AURKA, AXIN2, BAP1, BCL2, BCR, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRIP1, BTK, BUB IB, CALR, CBL, CCND1, CCNE1, CDC73, CDH1, CDK4, CDK6, CDKN1B, CDKN2A, CDKN2B, CDKN2C, CEBPA, CHEK2, CIC, CREBBP, CSF1R, CTNNB1, CYLD, DAXX, DDB2, DDR2, DICERl, DNMT3A, EGFR, EP300, ERBB2, ERBB3, ERBB4, ERCC1, ERCC2, ERCC3, ERCC4, ERCC5, ESR1, ETV1, ETV5, EWSR1, EXT1, EXT2, FANCA, FANCB, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCL FANCL, FANCM, FBXW7, FGFR1, FGFR2, FGFR3, FGFR4, FH, FLCN, FLT3, FLT4, FOXL2, GATA1, GATA2, GNA11, GNAQ, GNAS, GPC3, H3F3A, H3F3B, HNF1A, HRAS, IDH1, IDH2, IGF1R, IGF2R, KZFl, JAK1, JAK2, JAK3, KDR, KIT, KRAS, MAML1, MAP2K1, MAP2K4, MDM2, MDM4, MED12, MENl, MET, MLH1, MLL, MPL, MSH2, MSH6, MTOR, MUTYH, MYCLl, MYCN, NBN, NCOA3, NF1, NF2, NKX2-1, NOTCH1, NOTCH2, NOTCH3, NOTCH4, NPM1, NRAS, NTRK1, PALB2, PAX5, PBRMl, PDGFRA, PHOX2B, PIK3CA, PIK3R1, PMS1, PMS2, POLD1, POLE, POLH, POT1, PRKARIA, PRSS1, PTCH1, PTEN, PTPNl 1, RAD51C, RAF1, RBI, RECQL4, RET, RNF43, ROS1, RUNX1, SBDS, SDHAF2, SDHB, SDHC, SDHD, SF3B1, SMAD2, SMAD3, SMAD4, SMARCBl, SMO, SRC, STAG2, STK11, SUFU, TERT, TET2, TGFBR2, TNFAIP3, TOPI, TP53, TSC1, TSC2, TSHR, VHL, WAS, WRN, WT1, XPA, XPC, and/or XRCC1. In some embodiments, the subject has at least one mutation in one or more sequences encoding ABLl, ACVR1, AKT1, AKT2, ALK, APC, AR, ARIDIA, ARID IB, ASXL1, ATM, ATRX, AURKA, AXIN2, BAP1, BCL2, BCR, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRIP1, BTK, BUB IB, CALR, CBL, CCND1, CCNE1, CDC73, CDH1, CDK4, CDK6, CDKN1B, CDKN2A, CDKN2B, CDKN2C, CEBPA, CHEK2, CIC, CREBBP, CSF1R, CTNNB1, CYLD, DAXX, DDB2, DDR2, DICERl, DNMT3A, EGFR, EP300, ERBB2, ERBB3, ERBB4, ERCC1, ERCC2, ERCC3, ERCC4, ERCC5, ESR1, ETVl, ETV5, EWSR1, EXTl, EXT2, EZH2, FANCA, FANCB, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCL, FANCM, FBXW7, FGFR1, FGFR2, FGFR3, FGFR4, FH, FLCN, FLT3, FLT4, FOXL2, GATA1, GATA2, GNA11, GNAQ, GNAS, GPC3, H3F3A, H3F3B, HNFIA, HRAS, IDH1, IDH2, IGF1R, IGF2R, IKZF1, JAK1, JAK2, JAK3, KDR, KIT, KRAS, MAMLl, MAP2K1, MAP2K4, MDM2, MDM4, MED 12, MENl, MET, MLHl, MLL, MPL, MSH2, MSH6, MTOR, MUTYH, MYCLl, MYCN, NBN, NCOA3, NFl, NF2, NKX2-1, NOTCH1, NOTCH2, NOTCH3, NOTCH4, NPM1, NRAS, NTRK1, PALB2, PAX5, PBRMl, PDGFRA, PHOX2B, PIK3CA, PIK3R1, PMS1, PMS2, POLD1, POLE, POLH, POT1, PRKARIA, PRSS1, PTCH1, PTEN, PTPNl 1, RAD51C, RAF1, RBI, RECQL4, RET, RNF43, ROS1, RUNX1, SBDS, SDHAF2, SDHB, SDHC, SDHD, SF3B1, SMAD2, SMAD3, SMAD4, SMARCBl, SMO, SRC, STAG2, STK11, SUFU, TERT, TET2, TGFBR2, TNFAIP3, TOPI, TP53, TSC1, TSC2, TSHR, VHL, WAS, WRN, WT1, XPA, XPC, and/or XRCC1. In some embodiments, the subject has at least one mutation in one or more sequences encoding ARID1 A, ATM, B2M, BCL2, BCL6, BCL7A, BRAF, BTG1, CARD11, CCND3, CD58, CD79B, CDKN2A, CREBBP, EP300, EZH2, FOXOl, GNA13, fflSTlHIB, fflSTlHIC, HIST1H1E, IKZF3, IRF4, ΓΓΡΚΒ, KDM6A, KIT, KMT2D, KRAS, MEF2B, MYC, MYD88, NOTCH1, NOTCH2, NRAS, PIK3CA, PIM1, POU2F2, PRDM1, PTEN, PTPN1, PTPN11, PTPN6, PTPRD, RBI, S1PR2, SGK1, SMARCB1, SOCS1, STAT6, TBL1XR1, TNFAIP3, TNFRSF14, TP53, XPOl. In some embodiments, the subject has at least one mutation in one or more sequences encoding AKTl, ALK, ARID 1 A, ATM, B2M, BCL2, BCL6, BCL7A, BTG2, CARDll, CCND3, CD79B, CDKN2A, CREBBP, EP300, EZH2, FBXW7, FOXOl, HLA-C, HRAS, IKZF3, IRF4, KDM6A, KRAS, MEF2B, MYD88, NOTCH1, NPM1, NRAS, PIK3CA, PIM1, PRDMl, PTEN, RBI, RBBP4,
SMARCBl, SUZ12, TNFRSF14, and/or TP53. In some embodiments, the subject has at least one mutation in one or more sequences encoding ALK, EWSR1, ROS1, BCL2, MLL,
TMPRSS2, BCR, MYC, FGFR3, BRAF, NTRKl, TACC3, DNAJB1, PDGFRA, EGFR, PDGFRB, ETV1, PRKACA, ETV4, RAFl, ETV5, RARA, ETV6, RET. In some embodiments, the subject has at least one mutation in one or more sequences encoding ALK (Intron 19), BCL2 (MBR breakpoint region), BCL2 (MCR breakpoint region), BCL6, CD274, CIITA, MYC (entire Gene + 40kbp upstream), and/or PDCD1 LG2. In some embodiments, the subject has at least one mutation in one or more sequences encoding BCL2, CD274 (PDL1), FOXP1, JAK2, KDM4C, PDCD1LG2 (PDL2), and/or REL. In some embodiments, the subject has at least one mutation in one or more sequences encoding ARIDl A, ATM, B2M, BCL2, BCL6, BCL7A, BRAF,
CARDll, CCND3, CD274 (PDL1), CD58, CD79B, CDKN2A, CIITA, CREBBP, EZH2 (non- Y646), EZH2 (Y646), EP300, FOXOl, FOXP1, GNA13, fflSTlHIB, fflSTlHIC, fflSTlHlE, IRF4, IZKF3, JAK2, KDM4C, KDM6A, KIT, KMT2D, KRAS, MEF2B, MYC, MYD88, NOTCH1, NOTCH2, NRAS, PDCD1LG2 (PDL2), PIK3CA, PIM1, POU2F2, PRDMl, PTEN, PTPN11, PTPN6, PTPRD, REL, SOCS1, STAT6, TNFAIP3, TNFRSF14, and/or TP53. In some embodiments, the subject has at least one mutation in one or more sequences encoding
ARIDl A, B2M, BCL2, BCL6, CARDl 1, CCND3, CD274 (PDL1), CD58, CD79B, CDKN2A, CREBBP, EZH2, EP300, FOXOl, GNA13, HIST1H1B, fflSTlHIC, fflSTlHlE, KMT2D, KRAS, MEF2B, MYC, MYD88 (273P), PDCD1LG2 (PDL2), PIM1, POU2F2, PRDMl, SOCS1, STAT6, TNFAIP3, and/or TNFRSF14. In some embodiments, the subject has at least one mutation in in one or more sequences encoding: EZH2, MYD88, STAT6A, SOCS1, MYC, and/or fflSTlHlE, [070J In some embodiments, the subject has at least one mutation that decreases or abolishes the function of a gene product (e.g., a transcript, mRNA, or protein) encoded by the mutated sequence as compared to the function of the respective gene product encoded by the wild-type sequence. Such mutations are also sometimes referred to as loss-of-function mutations. Many loss-of-function mutations for the genes and gene products referred to herein that are suitable for some embodiments of this disclosure will be known to the skilled artisan. For example, in some exemplary embodiments, the subject has a loss-of-function mutation in SOCS1. In some embodiments, the subject has at least one mutation that increases the function of a gene product (e.g., a transcript, mRNA, or protein) encoded by the mutated sequence as compared to the function of the respective gene product encoded by the wild-type sequence. Such mutations are also sometimes referred to as gain-of-function mutations or activating mutations. Many gain-of- function mutations for the genes and gene products referred to herein that are suitable for some embodiments of this disclosure will be known to the skilled artisan. For example, in some embodiments, the subject has a gain-of-function mutation in a sequence encoding EZH2, MYD88, STAT6, or MYC. In some embodiments, the subject has at least one loss-of-function and at least one gain-of function mutation. For example, in some embodiments, the subject has at least one gain-of-function mutation in a sequence encoding EZH2 or STAT6, and at least one loss-of-function mutation in a sequence encoding SOCS1. In some embodiments, the subject does not have a specific mutation, e.g., a gain-of-function in a sequence encoding MYC or a loss-of-function mutation in SOCS1.
[071] In some embodiments, the subject expresses a mutant EZH2 protein. In some embodiments, the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1, a substitution of any amino acid other than alanine (A) for alanine (A) at position 682 of SEQ ID NO: 1, and/or a substitution of any amino acid other than alanine (A) for alanine (A) at position 692 of SEQ ID NO: 1. In some embodiments, the subject expresses at least one mutant MYD88, STAT6, and/or a SOCS1 protein, either in addition to the mutant EZH2 protein or in the absence of a mutant EZH2 protein. In some embodiments, the subject does not express a mutant MYC and/or a mutant HIST1H1E protein. In some embodiments, the mutant EZH2 protein, the mutant MYD88 protein, the mutant STAT6 protein, and/or the mutant MYC protein exhibits an increase in activity as compared to the respective wild-type protein. In some embodiments, the mutant SOCS1 protein exhibits a decreased activity as compared to the respective wild-type SOCS1 protein.
[072] In some embodiments, the methods provided herein further comprise detecting the at least one mutation in the subject. Such detecting may, in some embodiments, comprise subjecting a sample obtained from the subject to a suitable sequence analysis assay, e.g., to a next generation sequencing assay. Suitable sequencing assays are provided herein or otherwise known to those of skill in the art, and the disclosure is not limited in this respect.
[073] Some aspects of this disclosure provide methods comprising selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of at least one mutation associated with a positive response to such treatment in the subject and/or based on the absence of at least one mutation associated with no response or with a negative response to such treatment in the subject. In some embodiments, the at least one mutation associated with a positive response comprises (a) an EZH2 mutation (e.g., a gain-of-function EZH2 mutation); (b) a histone acetyl transferase (HAT) mutation; (c) a STAT6 mutation (e.g., a gain-of-function STAT6 mutation); (d) a MYD88 mutation (e.g., a gain-of-function MYD88 mutation); and/or (e) a SOCS1 mutation (e.g., a loss-of-function SOCS1 mutation). In some embodiments, the at least one mutation associated with no response or with a negative response comprises (a) a MYC mutation (e.g., a gain-of-function MYC mutation); and/or (b) a HISTIHIE mutation. In some embodiments, the method comprises detecting the at least one mutation associated with a positive response and/or the at least one mutation associated with no response or a negative response in a sample obtained from the subject by subjecting the sample to a suitable sequence analysis assay. In some embodiments, the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having at least one of a MYD88 mutation, a STAT6A mutation, and a SOCS1 mutation, and/or (b) not having at least one of a MYC mutation and/or a HISTIHIE mutation. In some embodiments, the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having at least one of a MYD88 mutation, a STAT6A mutation, and a SOCS1 mutation, and (b) not having a MYC mutation and a HISTIHIE mutation.
[074] Some aspects of this disclosure provide methods for selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of a mutation profile in the subject that matches a mutation profile (e.g., at least 2, at least 3, at least 4, or at least S, or more mutations, or, in some embodiments, all mutations), of a patient exhibiting a complete or partial response or stable disease as described in any of Figures 1-3.
Definitions
[075] According to the methods of the disclosure, a "normal" cell may be used as a basis of comparison for one or more characteristics of a cancer cell, including the presence of one or more mutations in a hi stone acetyltransferase that result in a decreased activity of the enzyme. For example, the one or more mutations in a histone acetyltransferase may result in a decreased acetylation activity or efficacy of the enzyme, and, consequently, a reduced or decreased level of acetylation of at least one lysine on Histone 3 (H3). In certain embodiments, the one or more mutations in a histone acetyltransferase may result in a decreased acetylation activity or efficacy of the enzyme, and, consequently, a reduced or decreased level of acetylation of lysine 27 on Histone 3 (H3) (H3K27). As used herein, a "normal cell" is a cell that cannot be classified as part of a "cell proliferative disorder". A normal cell lacks unregulated or abnormal growth, or both, that can lead to the development of an unwanted condition or disease. Preferably, a normal cell expresses a comparable amount of EZH2 as a cancer cell. Preferably a normal cell contains a wild type sequence for all histone acetyltransferases, expresses a histone
acetyltransferase transcript without mutations, and expresses a histone acetyltransferase protein without mutations that retains all functions a normal activity levels.
[076] As used herein, "contacting a cell" refers to a condition in which a compound or other composition of matter is in direct contact with a cell, or is close enough to induce a desired biological effect in a cell.
[077] As used herein, "treating" or "treat" describes the management and care of a subject for the purpose of combating a disease, condition, or disorder and includes the administration of an EZH2 inhibitor of the disclosure, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph or solvate thereof, to alleviate the symptoms or complications of cancer or to eliminate the cancer.
[078] As used herein, the term "alleviate" is meant to describe a process by which the severity of a sign or symptom of cancer is decreased. Importantly, a sign or symptom can be alleviated without being eliminated. In a preferred embodiment, the administration of pharmaceutical compositions of the disclosure leads to the elimination of a sign or symptom, however, elimination is not required. Effective dosages are expected to decrease the severity of a sign or symptom. For instance, a sign or symptom of a disorder such as cancer, which can occur in multiple locations, is alleviated if the severity of the cancer is decreased within at least one of multiple locations.
[079] As used herein, the term "severity" is meant to describe the potential of cancer to transform from a precancerous, or benign, state into a malignant state. Alternatively, or in addition, severity is meant to describe a cancer stage, for example, according to the TNM system
(accepted by the International Union Against Cancer (UICC) and the American Joint Committee on Cancer (AJCC)) or by other art-recognized methods. Cancer stage refers to the extent or severity of the cancer, based on factors such as the location of the primary tumor, tumor size, number of tumors, and lymph node involvement (spread of cancer into lymph nodes).
Alternatively, or in addition, severity is meant to describe the tumor grade by art-recognized methods (see, National Cancer Institute, www.cancer.gov). Tumor grade is a system used to classify cancer cells in terms of how abnormal they look under a microscope and how quickly the tumor is likely to grow and spread. Many factors are considered when determining tumor grade, including the structure and growth pattern of the cells. The specific factors used to determine tumor grade vary with each type of cancer. Severity also describes a histologic grade, also called differentiation, which refers to how much the tumor cells resemble normal cells of the same tissue type (see, National Cancer Institute, www.cancer.gov). Furthermore, severity describes a nuclear grade, which refers to the size and shape of the nucleus in tumor cells and the percentage of tumor cells that are dividing (see, National Cancer Institute, www. cancer.gov).
[080] In another aspect of the disclosure, severity describes the degree to which a tumor has secreted growth factors, degraded the extracellular matrix, become vascularized, lost adhesion to juxtaposed tissues, or metastasized. Moreover, severity describes the number of locations to which a primary tumor has metastasized. Finally, severity includes the difficulty of treating tumors of varying types and locations. For example, inoperable tumors, those cancers which have greater access to multiple body systems (hematological and immunological tumors), and those which are the most resistant to traditional treatments are considered most severe. In these situations, prolonging the life expectancy of the subject and/or reducing pain, decreasing the proportion of cancerous cells or restricting cells to one system, and improving cancer stage/tumor grade/histological grade/nuclear grade are considered alleviating a sign or symptom of the cancer. [081J As used herein the term "symptom" is defined as an indication of disease, illness, injury, or that something is not right in the body. Symptoms are felt or noticed by the individual experiencing the symptom, but may not easily be noticed by others. Others are defined as non- health-care professionals.
[082] As used herein the term "sign" is also defined as an indication that something is not right in the body. But signs are defined as things that can be seen by a doctor, nurse, or other health care professional.
[083] Cancer is a group of diseases that may cause almost any sign or symptom. The signs and symptoms will depend on where the cancer is, the size of the cancer, and how much it affects the nearby organs or structures. If a cancer spreads (metastasizes), then symptoms may appear in different parts of the body.
[084] As a cancer grows, it begins to push on nearby organs, blood vessels, and nerves. This pressure creates some of the signs and symptoms of cancer. Cancers may form in places where it does not cause any symptoms until the cancer has grown quite large.
[085] Cancer may also cause symptoms such as fever, fatigue, or weight loss. This may be because cancer cells use up much of the body's energy supply or release substances that change the body's metabolism. Or the cancer may cause the immune system to react in ways that produce these symptoms. While the signs and symptoms listed above are the more common ones seen with cancer, there are many others that are less common and are not listed here. However, all art-recognized signs and symptoms of cancer are contemplated and encompassed by the disclosure.
[086] Treating cancer may result in a reduction in size of a tumor. A reduction in size of a tumor may also be referred to as "tumor regression". Preferably, after treatment according to the methods of the disclosure, tumor size is reduced by 5% or greater relative to its size prior to treatment; more preferably, tumor size is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75% or greater. Size of a tumor may be measured by any reproducible means of measurement. The size of a tumor may be measured as a diameter of the tumor.
[087] Treating cancer may result in a reduction in tumor volume. Preferably, after treatment according to the methods of the disclosure, tumor volume is reduced by 5% or greater relative to its size prior to treatment; more preferably, tumor volume is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75% or greater. Tumor volume may be measured by any reproducible means of measurement.
[088] Treating cancer may result in a decrease in number of tumors. Preferably, after treatment, tumor number is reduced by 5% or greater relative to number prior to treatment; more preferably, tumor number is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75%. Number of tumors may be measured by any reproducible means of measurement. The number of tumors may be measured by counting tumors visible to the naked eye or at a specified magnification. Preferably, the specified magnification is 2x, 3x, 4x, 5x, 10x, or 50x.
[089] Treating cancer may result in a decrease in number of metastatic lesions in other tissues or organs distant from the primary tumor site. Preferably, after treatment according to the methods of the disclosure, the number of metastatic lesions is reduced by 5% or greater relative to number prior to treatment; more preferably, the number of metastatic lesions is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75%. The number of metastatic lesions may be measured by any reproducible means of measurement. The number of metastatic lesions may be measured by counting metastatic lesions visible to the naked eye or at a specified magnification. Preferably, the specified magnification is 2x, 3x, 4x, 5x, lOx, or 50x.
[090] An effective amount of an EZH2 inhibitor of the disclosure, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph or solvate thereof, is not significantly cytotoxic to normal cells. For example, a therapeutically effective amount of an EZH2 inhibitor of the disclosure is not significantly cytotoxic to normal cells if administration of the EZH2 inhibitor of the disclosure in a therapeutically effective amount does not induce cell death in greater than 10% of normal cells. A therapeutically effective amount of an EZH2 inhibitor of the disclosure does not significantly affect the viability of normal cells if administration of the compound in a therapeutically effective amount does not induce cell death in greater than 10% of normal cells.
[091] Contacting a cell with an EZH2 inhibitor of the disclosure, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph or solvate thereof, can inhibit EZH2 activity selectively in cancer cells. Administering to a subject in need thereof an EZH2 inhibitor of the disclosure, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph or solvate thereof, can inhibit EZH2 activity selectively in cancer cells.
EZH2 Inhibitors
[092J EZH2 inhibitors of the disclosure comprise tazemetostat (EPZ-6438):
Figure imgf000049_0001
or a pharmaceutically acceptable salt thereof.
[093] Tazemetostat is also described in US Patent Nos. 8,410,088, 8,765,732, and 9,090,562 (the contents of which are each incorporated herein in their entireties).
[094] Tazemetostat or a pharmaceutically acceptable salt thereof, as described herein, is potent in targeting both WT and mutant EZH2. Tazemetostat is orally bioavailable and has high selectivity to EZH2 compared with other histone methyltransferases (i.e., >20,000 fold selectivity by Ki). Importantly, tazemetostat has targeted methyl mark inhibition that results in the killing of genetically defined cancer cells in vitro. Animal models have also shown sustained in vivo efficacy following inhibition of the target methyl mark. Clinical trial results described herein also demonstrate the safety and efficacy of tazemetostat.
[095] In some embodiments, tazemetostat or a pharmaceutically acceptable salt thereof is administered to the subject at a dose of approximately 100 mg to approximately 3200 mg daily, such as about 100 mg BID to about 1600 mg BID (e.g., 100 mg BID, 200 mg BID, 400 mg BID, 800 mg BID, or 1600 mg BID), for treating a NHL. On one embodiment the dose is 800 mg BID.
[096] EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of:
Figure imgf000050_0001
thereof or pharmaceutically acceptable salts and solvates thereof.
[097] EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of Compound E:
Figure imgf000050_0002
(E) or pharmaceutically acceptable salts thereof.
[098] EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of GSK-126, having the following formula:
Figure imgf000051_0001
stereoisomers thereof, or pharmaceutically acceptable salts or solvates thereof.
[099] EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of Compound F:
or stereoisomers thereof or pharmaceutically
Figure imgf000051_0002
acceptable salts and solvates thereof.
[0100] EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of any one of Compounds Ga-Gc:
Figure imgf000051_0003
or a stereoisomer, pharmaceutically
Figure imgf000052_0001
acceptable salt or solvate thereof.
[0101] EZH2 inhibitors of the disclosure may comprise, consist essentially of or consist of CPI-1205 or GSK343.
[0102] Additional suitable EZH2 inhibitors will be apparent to those skilled in the art. In some embodiments of the strategies, treatment modalities, methods, combinations, and compositions provided herein, the EZH2 inhibitor is an EZH2 inhibitor described in US 8,536,179 (describing GSK-126 among other compounds and corresponding to WO 2011/140324), the entire contents of each of which are incorporated herein by reference.
[0103] In some embodiments of the strategies, treatment modalities, methods, combinations, and compositions provided herein, the EZH2 inhibitor is an EZH2 inhibitor described in PCT/US2014/015706, published as WO 2014/124418, in PCT/US2013/025639, published as WO 2013/120104, and in US 14/839,273, published as US 2015/0368229, the entire contents of each of which are incorporated herein by reference.
[0104] In some embodiments, the compound disclosed herein is the compound itself, i.e., the free base or "naked" molecule. In some embodiments, the compound is a salt thereof, e.g., a mono-HCl or tri-HCl salt, mono-HBr or tri-HBr salt of the naked molecule.
[0105] Compounds disclosed herein that contain nitrogens can be converted to N-oxides by treatment with an oxidizing agent (e.g., 3-chloroperoxybenzoic acid (/HCPBA) and/or hydrogen peroxides) to afford other compounds suitable for any methods disclosed herein. Thus, all shown and claimed nitrogen-containing compounds are considered, when allowed by valency and structure, to include both the compound as shown and its N-oxide derivative (which can be designated as N DO or N+-0"). Furthermore, in other instances, the nitrogens in the compounds disclosed herein can be converted to N-hydroxy or N-alkoxy compounds. For example, N- hydroxy compounds can be prepared by oxidation of the parent amine by an oxidizing agent such as /H-CPBA. All shown and claimed nitrogen-containing compounds are also considered, when allowed by valency and structure, to cover both the compound as shown and its N- hydroxy (i.e., N-OH) and N-alkoxy (i.e., N-OR, wherein R is substituted or unsubstituted Ci-C 6 alkyl, Ci-Ce alkenyl, C1-C6 alkynyl, 3-14-membered carbocycle or 3-14-membered heterocycle) derivatives.
[0106] "Isomerism" means compounds that have identical molecular formulae but differ in the sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers." Stereoisomers that are not mirror images of one another are termed "diastereoisomers," and stereoisomers that are non-superimposable mirror images of each other are termed "enantiomers" or sometimes optical isomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a "racemic mixture."
[0107] A carbon atom bonded to four nonidentical substituents is termed a "chiral center."
[0108] "Chiral isomer" means a compound with at least one chiral center. Compounds with more than one chiral center may exist either as an individual diastereomer or as a mixture of diastereomers, termed "diastereomeric mixture." When one chiral center is present, a stereoisomer may be characterized by the absolute configuration (R or S) of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al., Angew. Chem. Inter. Edit. 1966, 5, 385; errata 511; Cahn etal., Angew. Chem. 1966, 78, 413; Cahn and Ingold, J. Chem. Soc. 1951 (London), 612; Cahn etal, Experientia 1956, 12, 81; Cahn, J. Chem. Educ. 1964, 41, 116).
[0109] "Geometric isomer" means the diastereomers that owe their existence to hindered rotation about double bonds or a cycloalkyl linker (e.g., 1, 3-cylcobutyl). These configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold-Prelog rules.
[0110] It is to be understood that the compounds disclosed herein may be depicted as different chiral isomers or geometric isomers. It should also be understood that when compounds have chiral isomeric or geometric isomeric forms, all isomeric forms are intended to be included in the scope of the disclosure, and the naming of the compounds does not exclude any isomeric forms. [0111] Furthermore, the structures and other compounds discussed in thi s di sclosure include all atropic isomers thereof. "Atropic isomers" are a type of stereoisomer in which the atoms of two isomers are arranged differently in space. Atropic isomers owe their existence to a restricted rotation caused by hindrance of rotation of large groups about a central bond. Such atropic isomers typically exist as a mixture, however as a result of recent advances in chromatography techniques; it has been possible to separate mixtures of two atropic isomers in select cases.
[0112] "Tautomer" is one of two or more structural isomers that exist in equilibrium and is readily converted from one isomeric form to another. This conversion results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. Tautomers exist as a mixture of a tautomeric set in solution. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent and pH. The concept of tautomers that are interconvertible by tautomerization is called tautomerism.
[0113] Of the various types of tautomerism that are possible, two are commonly observed. In keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs. Ring- chain tautomerism arises as a result of the aldehyde group (-CHO) in a sugar chain molecule reacting with one of the hydroxy groups (-OH) in the same molecule to give it a cyclic (ring- shaped) form as exhibited by glucose.
[0114] Common tautomeric pairs are: ketone-enol, amide-nitrile, lactam-lactim, amide-imidic acid tautomerism in heterocyclic rings (e.g., in nucleobases such as guanine, thymine and cytosine), imine-enamine and enamine-enamine. An example of keto-enol equilibria is between pyridin-2(lH)-ones and the corresponding pyridin-2-ols, as shown below.
Figure imgf000054_0001
[0115] It is to be understood that the compounds disclosed herein may be depicted as different tautomers. It should also be understood that when compounds have tautomeric forms, all tautomeric forms are intended to be included in the scope of the disclosure, and the naming of the compounds does not exclude any tautomer form. [0116] The compounds disclosed herein include the compounds themselves, as well as their salts and their solvates, if applicable. A salt, for example, can be formed between an anion and a positively charged group (e.g., amino) on an aryl- or heteroaryl-substituted benzene compound. Suitable anions include chloride, bromide, iodide, sulfate, bi sulfate, sulfamate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, glutamate, glucuronate, glutarate, malate, maleate, succinate, fumarate, tartrate, tosylate, salicylate, lactate, naphthalenesulfonate, and acetate (e.g., trifluoroacetate). The term "pharmaceutically acceptable anion" refers to an anion suitable for forming a pharmaceutically acceptable salt. Likewise, a salt can also be formed between a cation and a negatively charged group (e.g., carboxylate) on an aryl- or heteroaryl-substituted benzene compound. Suitable cations include sodium ion, potassium ion, magnesium ion, calcium ion, and an ammonium cation such as tetramethylammonium ion. The aryl- or heteroaryl-substituted benzene compounds also include those salts containing quaternary nitrogen atoms. In the salt form, it is understood that the ratio of the compound to the cation or anion of the salt can be 1:1, or any ration other than 1:1, e.g., 3:1, 2:1, 1:2, or 1:3.
[0117] Additionally, the compounds disclosed herein, for example, the salts of the compounds, can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules. Nonlimiting examples of hydrates include monohydrates, dihydrates, etc.
Nonlimiting examples of solvates include ethanol solvates, acetone solvates, etc.
[0118] "Solvate" means solvent addition forms that contain either stoichiometric or non- stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate; and if the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one molecule of the substance in which the water retains its molecular state as EbO.
[0119] As used herein, the term "analog" refers to a chemical compound that is structurally similar to another but differs slightly in composition (as in the replacement of one atom by an atom of a different element or in the presence of a particular functional group, or the
replacement of one functional group by another functional group). Thus, an analog is a compound that is similar or comparable in function and appearance, but not in structure or origin to the reference compound.
[0120] As defined herein, the term "derivative" refers to compounds that have a common core structure, and are substituted with various groups as described herein. For example, all of the compounds represented by Formula (I) are aryl- or heteroaryl-substituted benzene compounds, and have Formula (I) as a common core.
[0121] The term "bioisostere" refers to a compound resulting from the exchange of an atom or of a group of atoms with another, broadly similar, atom or group of atoms. The objective of a bioisosteric replacement is to create a new compound with similar biological properties to the parent compound. The bioisosteric replacement may be physicochemically or topological^ based. Examples of carboxylic acid bioisosteres include, but are not limited to, acyl
sulfonimides, tetrazoles, sulfonates and phosphonates. See, e.g., Patani and LaVoie, Chem. Rev. 96, 3147-3176, 1996.
[0122] The present disclosure is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium, and isotopes of carbon include C-13 and C-14.
Pharmaceutical Formulations
[0123] The present disclosure also provides pharmaceutical compositions comprising at least one EZH2 inhibitor described herein in combination with at least one pharmaceutically acceptable excipient or carrier.
[0124] A "pharmaceutical composition" is a formulation containing the EZH2 inhibitors of the present disclosure in a form suitable for administration to a subject. In some embodiments, the pharmaceutical composition is in bulk or in unit dosage form. The unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler or a vial. The quantity of active ingredient {e.g., a formulation of the disclosed compound or salt, hydrate, solvate or isomer thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved. One skilled in the art will appreciate that it is sometimes necessary to make routine variations to the dosage depending on the age and condition of the patient. The dosage will also depend on the route of administration. A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like. Dosage forms for the topical or transdermal administration of a compound of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. In some embodiments, the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers or propellants that are required.
[0125] As used herein, the phrase "pharmaceutically acceptable" refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[0126] "Pharmaceutically acceptable excipient" means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use. A "pharmaceutically acceptable excipient" as used in the disclosure includes both one and more than one such excipient.
[0127] A pharmaceutical composition of the disclosure is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), and transmucosal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens;
antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as
ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
[0128] A compound or pharmaceutical composition of the disclosure can be administered to a subject in many of the well-known methods currently used for chemotherapeutic treatment. For example, for treatment of cancers, a compound of the disclosure may be injected directly into tumors, injected into the blood stream or body cavities or taken orally or applied through the skin with patches. The dose chosen should be sufficient to constitute effective treatment but not as high as to cause unacceptable side effects. The state of the disease condition (e.g., cancer, precancer, and the like) and the health of the patient should preferably be closely monitored during and for a reasonable period after treatment. [0129] The term "therapeutically effective amount", as used herein, refers to an amount of an EZH2 inhibitor, composition, or pharmaceutical composition thereof effective to treat, ameliorate, or prevent an identified disease or condition, or to exhibit a detectable therapeutic or inhibitory effect. The effect can be detected by any assay method known in the art. The precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration. Therapeutically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician. In a preferred aspect, the disease or condition to be treated is cancer, including but not limited to, B cell lymphoma, including activated B-cell (ABC) and germinal B-cell (GBC) subtypes.
[0130] For any EZH2 inhibitor of the disclosure, the therapeutically effective amount can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models, usually rats, mice, rabbits, dogs, or pigs. The animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. Therapeutic/prophylactic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., EDso (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LDso/EDso. Pharmaceutical compositions that exhibit large therapeutic indices are preferred. The dosage may vary within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
[0131] Dosage and administration are adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular formulation.
[0132] The pharmaceutical compositions containing an EZH2 inhibitor of the present disclosure may be manufactured in a manner that is generally known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes. Pharmaceutical compositions may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and/or auxiliaries that facilitate processing of the active compounds into preparations that can be used pharmaceutically. Of course, the appropriate formulation is dependent upon the route of administration chosen.
[0133] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
[0134] Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. [0135] Oral compositions generally include an inert diluent or an edible pharmaceutically acceptable carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
[0136] For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser, which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
[0137] Sy stemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
[0138] The active compounds (e.g., EZH2 inhibitors of the disclosure) can be prepared with pharmaceutically acceptable carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and
microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
[0139] It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the disclosure are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.
[0140] In therapeutic applications, the dosages of the pharmaceutical compositions used in accordance with the disclosure vary depending on the agent, the age, weight, and clinical condition of the recipient patient, and the experience and judgment of the clinician or practitioner administering the therapy, among other factors affecting the selected dosage.
Generally, the dose should be sufficient to result in slowing, and preferably regressing, the growth of the tumors and also preferably causing complete regression of the cancer. An effective amount of a pharmaceutical agent is that which provides an objectively identifiable improvement as noted by the clinician or other qualified observer. For example, regression of a tumor in a patient may be measured with reference to the diameter of a tumor. Decrease in the diameter of a tumor indicates regression. Regression is also indicated by failure of tumors to reoccur after treatment has stopped. As used herein, the term "dosage effective manner" refers to amount of an active compound to produce the desired biological effect in a subject or cell.
[0141] The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
[0142] The compounds of the present disclosure are capable of further forming salts. All of these forms are also contemplated within the scope of the claimed disclosure.
[0143] As used herein, "pharmaceutically acceptable salts" refer to derivatives of the compounds of the present disclosure wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines, alkali or organic salts of acidic residues such as carboxylic acids, and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include, but are not limited to, those derived from inorganic and organic acids selected from 2-acetoxybenzoic, 2-hydroxyethane sulfonic, acetic, ascorbic, benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edetic, ethane disulfonic, 1,2-ethane sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, glycollyarsanilic,
hexylresorcinic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxymaleic, hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic, malic, mandelic, methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic, phenylacetic, phosphoric, polygalacturonic, propionic, salicyclic, stearic, sub acetic, succinic, sulfamic, sulfanilic, sulfuric, tannic, tartaric, toluene sulfonic, and the commonly occurring amine acids, e.g., glycine, alanine, phenylalanine, arginine, etc.
[0144] Other examples of pharmaceutically acceptable salts include hexanoic acid, cyclopentane propionic acid, pyruvic acid, malonic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo-[2.2.2]-oct-2-ene-l-carboxylic acid, 3- phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, muconic acid, and the like. The present disclosure also encompasses salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine,
diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
[0145] It should be understood that all references to pharmaceutically acceptable salts include solvent addition forms (solvates) or crystal forms (polymorphs) as defined herein, of the same salt.
[0146] The EZH2 inhibitors of the present disclosure can also be prepared as esters, for example, pharmaceutically acceptable esters. For example, a carboxylic acid function group in a compound can be converted to its corresponding ester, e.g., a methyl, ethyl or other ester. Also, an alcohol group in a compound can be converted to its corresponding ester, e.g., an acetate, propionate or other ester.
[0147] The EZH2 inhibitors of the present disclosure can also be prepared as prodrugs, for example, pharmaceutically acceptable prodrugs. The terms "pro-drug" and "prodrug" are used interchangeably herein and refer to any compound which releases an active parent drug in vivo. Since prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, etc.), the compounds of the present disclosure can be delivered in prodrug form. Thus, the present disclosure is intended to cover prodrugs of the presently claimed compounds, methods of delivering the same and compositions containing the same. "Prodrugs" are intended to include any covalently bonded carriers that release an active parent drug of the present disclosure in vivo when such prodrug is administered to a subject. Prodrugs in the present disclosure are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Prodrugs include compounds of the present disclosure wherein a hydroxy, amino, sulfhydryl, carboxy or carbonyl group is bonded to any group that may be cleaved in vivo to form a free hydroxyl, free amino, free sulfhydryl, free carboxy or free carbonyl group, respectively.
[0148] Examples of prodrugs include, but are not limited to, esters (e.g., acetate,
dialkylaminoacetates, formates, phosphates, sulfates and benzoate derivatives) and carbamates (e.g., N,N-dimethylaminocarbonyl) of hydroxy functional groups, esters (e.g., ethyl esters, morpholinoethanol esters) of carboxyl functional groups, N-acyl derivatives (e.g., N-acetyl) N- Mannich bases, Schiff bases and enaminones of amino functional groups, oximes, acetals, ketals and enol esters of ketone and aldehyde functional groups in compounds of the disclosure, and the like, See Bundegaard, H., Design of Prodrugs, pl-92, Elesevier, New York-Oxford (1985).
[0149] The EZH2 inhibitors, or pharmaceutically acceptable salts, esters or prodrugs thereof, are administered orally, nasally, transdermally, pulmonary, inhalationally, buccally,
sublingually, intraperitoneally, subcutaneously, intramuscularly, intravenously, rectally, intrapleurally, intrathecally and parenterally. In some embodiments, the compound is administered orally. One skilled in the art will recognize the advantages of certain routes of administration.
[0150] The dosage regimen utilizing the compounds is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed. An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
[0151] The dosage regimen can be daily administration (e.g., every 24 hours) of a compound of the present disclosure. The dosage regimen can be daily administration for consecutive days, for example, at least two, at least three, at least four, at least five, at least six or at least seven consecutive days. Dosing can be more than one time daily, for example, twice, three times or four times daily (per a 24 hour period). The dosing regimen can be a daily administration followed by at least one day, at least two days, at least three days, at least four days, at least five days, or at least six days, without administration.
[0152] Techniques for formulation and administration of the disclosed compounds of the disclosure can be found in Remington: the Science and Practice of Pharmacy, 19th edition, Mack Publishing Co., Easton, PA (1995). In some embodiments, the compounds described herein, and the pharmaceutically acceptable salts thereof, are used in pharmaceutical
preparations in combination with a pharmaceutically acceptable carrier or diluent. Suitable pharmaceutically acceptable carriers include inert solid fillers or diluents and sterile aqueous or organic solutions. The compounds will be present in such pharmaceutical compositions in amounts sufficient to provide the desired dosage amount in the range described herein.
[0153] Methods of the disclosure for treating cancer including treating a B cell lymphoma, including the activated B-cell (ABC) and germinal B-cell (GBC) subtypes. In preferred embodiments, methods of the disclosure are used to treat a subject having a B cell lymphoma. In certain embodiments, the B cell lymphoma cell and/or the subject are characterized as having one or more mutations in a sequence that encodes a histone acetyltransferase (HAT). B cell lymphoma cells may contain a mutation in a gene that encodes a HAT, a corresponding HAT transcript (or cDNA copy thereof), or a HAT protein that decreases/inhibits an activity of a HAT protein. In preferred embodiments, the mutation in a gene that encodes a HAT, a corresponding HAT transcript (or cDNA copy thereof), or a HAT protein that decreases/inhibits an activity of a HAT protein, decreases or inhibits an acetylation activity or efficacy of the enzyme, resulting in a decreased level of acetylation of one or more lysines of histone 3 (H3) (e.g., H3K27). The presence of the HAT mutation resulting in a decreased level of acetylation of one or more lysines of histone 3 (H3) (e.g., H3K27) in a cell renders that cell sensitive to oncogenic transformation and treatment with an EZH2 inhibitor.
[0154] Methods of the disclosure may be used to treat a subject who has one or more mutations in a HAT that decrease/inhibit the ability of the HAT to acetylate one or more lysines of histone 3 (H3) (e.g., H3K27) or who has one or more cells with one or more mutations in a HAT that decrease/inhibit the ability of the HAT to acetylate one or more lysines of histone 3 (H3) (e.g., H3K27). HAT expression and/or HAT function may be evaluated by fluorescent and non- fluorescent immunohistochemistry (IHC) methods, including well known to one of ordinary skill in the art. In a certain embodiment the method comprises: (a) obtaining a biological sample from the subject; (b) contacting the biological sample or a portion thereof with an antibody that specifically binds HAT; and (c) detecting an amount of the antibody that is bound to HAT. Alternatively, or in addition, HAT expression and/or HAT function may be evaluated by a method comprising: (a) obtaining a biological sample from the subject; (b) sequencing at least one DNA sequence encoding a HAT protein from the biological sample or a portion thereof; and (c) determining if the at least one DNA sequence encoding a HAT protein contains a mutation affecting the expression and/or function of the HAT protein. HAT expression or a function of HAT may be evaluated by detecting an amount of the antibody that is bound to HAT and by sequencing at least one DNA sequence encoding a HAT protein, optionally, using the same biological sample from the subject.
[01SS] All percentages and ratios used herein, unless otherwise indicated, are by weight.
[0156] Other features and advantages of the present disclosure are apparent from the different examples. The provided examples illustrate different components and methodology useful in practicing the present disclosure. The examples do not limit the claimed disclosure. Based on the present disclosure the skilled artisan can identify and employ other components and methodology useful for practicing the present disclosure.
EXAMPLES
[0157] In order that the invention disclosed herein may be more efficiently understood, examples are provided below. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting the disclosure in any manner.
Example 1: Molecular Predictors of Response to EZH2 inhibition in NHL patients
[0158] Some B-cell malignancies depend on enhancer of zeste homolog 2 (EZH2; a hi stone methyl transferase) to perpetuate a less differentiated state, for example, on activating mutations of EZH2 being potential oncogenic drivers.
[0159] Tazemetostat is an oral, first-in-class, potent, and highly selective inhibitor of EZH2 currently in phase 2 clinical development for the treatment of relapsed or refractory non- Hodgkin lymphoma (NHL).
[0160] In the phase 1 part of a phase 1/2 study (NCT01897571) of adult patients with advanced solid tumors or B-cell lymphomas, objective responses to tazemetostat were observed in patients with tumors exhibiting mutated or wild-type EZH2.
[0161] An ongoing phase 2 study (NCT01897571) is enrolling patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) or follicular lymphoma (FL) to assess the efficacy and safety of tazemetostat. Patients with tumors exhibiting mutated or wild-type EZH2 are eligible to participate. The primary study endpoint is objective overall response rate.
[0162] A molecular analysis of tumor material obtained in the phase 2 study and its associations with preliminary response data is described herein, including, inter alia, a molecular predictor and a molecular predictor profile for responsiveness of a tumor or subject to treatment with an EZH2 inhibitor, e.g., with tazemetostat.
[0163] Methods & Results
[0164] During screening, archive tumor- and/or plasma-derived circulating tumor DNA (ctDNA) samples were obtained. Archive tumor was analyzed prospectively for EZ IE- activating mutations Y646F, Y646N, Y646X (Y646H, Y646C, and Y646S), A682G, and A692V using a cobas® EZH2 Mutation Test (Roche Molecular Systems, Inc., Pleasanton, CA, USA). Next generation sequencing (NGS) was performed retrospectively on DNA derived from archive tumor and plasma samples to identify somatic mutations, amplifications, and translocations using an NHL-specific 62-gene panel on the Alumina HiSeq 2S00 platform (Illumina, Inc., San
Diego, CA, USA) with 100 bp paired-end reads (Table 1).
Table i. Content for a targeted NGS panel representing 62 genes reported
to be mutated in >S% DL8CL or ft patients
Figure imgf000067_0001
[0165] Average target coverage for the tissue panel was 1250 X, while coverage for the ctDNA was approximately 20,000 X for mutations and 3700 X for structural alterations. Sequencing data were aligned to the human reference sequence (hgl9) and analyzed using validated cancer genome analysis algorithms (Personal Genome Diagnostics, Baltimore, MD, USA). EZH2 mutation detection rates in archive tumor were -12% in DLBCL and 19% in FL in the total phase 2 cohort (Table 2), consistent with previous reports.
Figure imgf000068_0001
[0166] Regardless of the technology or sample type used, the concordance rate for detection of EZH2 status was >95%. NGS and cobas® Test of archive tumor samples was 98.9% concordant (n=92) with 11 EZH2 activating mutation cases detected (Table 3). Concordance of EZH2 status between archive tumor and ctDNA samples was 97% (n=125).
Figure imgf000069_0001
[0167] Mutations in EZH2, STAT6, or MYD88 in wild-type EZH2 patients were associated with response (P<0.1), whereas mutations in fflSTlHlE, TP53, or MYC (P<0.08) were associated with nonresponse (Table 4).
[0168] Activating mutations in EZH2 and MYD88 (e.g., MYD88 L273P) were mutually exclusive. Accordingly, MYD88 was identified as a biomarker for response to treatment with EZH2 inhibitors, e.g., with tazemetostat, in wild-type EZH2 patients.
Figure imgf000069_0002
[0169] Patients matching a multigene predictor consisting of wild-type MYC and/or HIST1H1E but with mutated STAT6 and/or MYD88 in archive tumor had an objective overall response rate of -58% (11/19), whereas patients who did not match this profile had an objective overall response rate of -19% (14/73), demonstrating the potential for these four genes to predict response to tazemetostat (Table 5).
Figure imgf000070_0001
[0170] The molecular genetic profiling of NHL patients described here identifies predictors and predictor profiles for the response to treatment with EZH2 inhibitors, e.g., with tazemetostat. In addition, the results disclosed herein demonstrate that plasma-based ctDNA screening is a viable method to identify NHL patients with mutations, e.g., with EZH2-activating mutations, in the absence of archive tumor samples
[0171] All publications and patent documents cited herein are incorporated herein by reference as if each such publication or document was specifically and individually indicated to be incorporated herein by reference. Citation of publications and patent documents is not intended as an admission that any is pertinent prior art, nor does it constitute any admission as to the contents or date of the same. The invention having now been described by way of written description, those of skill in the art will recognize that the invention can be practiced in a variety of embodiments and that the foregoing description and examples below are for purposes of illustration and not limitation of the claims that follow. Where names of cell lines, genes or proteins are used herein, abbreviations and names conform to the nomenclature of the American Type Culture Collection (ATCC) or the National Center for Biotechnology Information (NCBI), unless otherwise noted or evident from the context.
[0172] The invention can be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims

What is claimed is:
1. A method, comprising administering a therapeutically effective amount of an inhibitor of Enhancer to Zeste Homolog 2 (EZH2) sufficient to treat a cancer to a subject in need thereof, wherein the cancer is characterized by at least one mutation in one or more sequences encoding a gene or gene product listed in Tables 1-5, and/or Figures 1-3.
2. The method of 1, wherein the cancer is characterized by at least one mutation in one or more sequences encoding: EZH2, MYD88, STAT6A, MYC, TP53, and/or HIST1H1E.
3. The method of claim 1, wherein the cancer is characterized by a genotype that satisfies Genetic Profile 1 of Figure 3.
4. The method of claim 1, wherein the cancer is characterized by a genotype that satisfies Genetic Profile 2 of Figure 3.
5. The method of claim 1, wherein the cancer comprises a MYD88 gain-of-function mutation.
6. The method of claim 5, wherein the cancer does not comprise an EZH2 gain-of-function mutation.
7. The method of any one of claims 1-6, wherein the at least one mutation decreases the function of a protein encoded by the mutated sequence as compared to the function of the protein encoded by the wild-type sequence.
8. The method of any one of claims 1-7, wherein the at least one mutation is a loss-of-function mutation.
9. The method of any one of claims 1-8, wherein the method further comprises detecting the at least one mutation in the subject.
10. The method of claim 9, wherein the detecting comprises subjecting a sample obtained from the subject to a sequence analysis assay.
11. The method of claim 10, wherein the analysis assay is a cobas® assay.
12. The method of claim 10 or 11, wherein the detecting comprises obtaining a sample from the subject.
13. The method of claim 12, wherein the sample is a tumor sample or a sample that comprises a cancer cell.
14. The method of claim 12, wherein the sample is a sample comprising ctDNA.
15. The method of any one of claims 1-14, wherein the inhibitor of EZH2 is (tazemetostat), or a pharmaceutically-acceptable salt thereof.
16. The method of any one of claims 1-15, wherein the inhibitor of EZH2 is administered orally.
17. The method of claim 16, wherein the inhibitor of EZH2 is formulated as a tablet.
18. The method of any one of claims 1-17, wherein the therapeutically effective amount of the inhibitor of EZH2 is between 100 mg and 3200 mg per day.
19. The method of claim 18, wherein the therapeutically effective amount of the inhibitor of EZH2 is 100 mg, 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, 1400 mg, 1600 mg or 3200 mg per day.
20. The method of claim 19, wherein the therapeutically effective amount is 1600 mg per day.
21. The method of any one of claims 1-20, wherein the cancer expresses a wild type EZH2 protein and does not express a mutant EZH2 protein.
22. The method of any one of claims 1-20, wherein the cancer expresses a mutant EZH2 protein.
23. The method of claim 22, wherein the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1.
24. The method of claim 22 or 23, wherein the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 682 of SEQ ID NO: 1.
25. The method of any one of claims 22-24, wherein the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 692 of SEQ ID NO: 1.
26. The method of any one of claims 1-25, wherein the at least one mutation comprises a MYD88 and/or STAT6A mutation, preferably wherein the MYD88 mutation is a gain-of-function mutation, preferably a MYD88 L273 mutation.
27. The method of any one of claims 1-26, wherein the cancer does not have a MYC and/or a fflSTlHlE mutation.
28. The method of any one of claims 1-27, wherein the cancer (a) has a MYD88 gain-of-function or a STAT6A loss-of-function mutation, and (b) has neither a MYC nor a HIST1H1E mutation.
29. The method of any one of claims 1-27, wherein the cancer (a) has a MYD88 L273 mutation or a STAT6A loss-of-function mutation, (b) has neither a MYC nor a HIST1H1E mutation, and (c) does not have an EZH2 gain-of-function mutation.
30. The method of any one of claims 1-29, wherein the subject is a human subject.
31. The method of any one of claims 1-30, wherein the subject has cancer.
32. The method of claim 31, wherein the cancer is B-cell lymphoma.
33. The method of claim 32, wherein the B-cell lymphoma is an activated B-cell (ABC) type.
34. The method of claim 32, wherein the B-cell lymphoma is a germinal B-cell (GBC) type.
35. The method of claim 31, wherein the cancer is follicular lymphoma.
36. A method, comprising selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of at least one mutation associated with a positive response to such treatment in the subject and/or based on the absence of at least one mutation associated with no response or with a negative response to such treatment in the subject.
37. The method of claim 36, wherein the at least one mutation associated with a positive response comprise (a) an EZH2 gain-of-function mutation or a MYD88 gain-of-function mutation; and, (b) a STAT6 loss-of-function mutation.
38. The method of claim 36 or 37, wherein the at least one mutation associated with no response or with a negative response comprise
(a) a MYC mutation;
(b) a TP53 mutation; and/or,
(c) a fflSTlHlE mutation.
39. The method of any one of claims 36-38, wherein the method comprises detecting the at least one mutation associated with a positive response and/or the at least one mutation associated with no response or a negative response in a sample obtained from the subject.
40. The method of any one of claims 36-39, wherein the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject (a) having either an EZH2 or a MYD88 gain-of-function mutation, or a STAT6A loss-of-function mutation; and (b) having neither a MYC mutation, nor a TP53 mutation, nor a HIST 1 HIE mutation.
41. The method of any one of claims 36-39, wherein the method comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject
(a) having a MYD88 L273 mutation or a STAT6A mutation; and,
(b) having neither a MYC mutation, nor a TP53 mutation, nor a HIST1H1E mutation.
42. The method of any one of claims 1-41, wherein the at least one mutation consists of a single mutation.
43. The method of any one of claims 1-41, wherein the at least one mutation compri ses 2 mutations or more.
44. The method of any one of claims 1-43, wherein the atleast one mutation comprises 3 mutations or more.
45. The method of any one of claims 1-44, wherein the at least one mutation comprises 4 mutations or more.
46. A method, comprising selecting a subject having cancer for treatment with an EZH2 inhibitor based on the presence of a mutation profile in the subject that matches a mutation profile of a patient exhibiting a complete or partial response or stable disease in any of Tables 1-5 or Figures 1-3.
47. Use of an inhibitor of Enhancer to Zeste Homolog 2 (EZH2) for treating a cancer in a subject in need thereof, the use comprising administering a therapeutically effective amount of the inhibitor to the subject, wherein the cancer is characterized by at least one mutation in one or more sequences encoding a gene or gene product listed in Tables 1-5, and/or Figures 1-3.
48. The use of claim 47, wherein the cancer is characterized by at least one mutation in one or more sequences encoding: EZH2, MYD88, STAT6A, MYC, TP53, and/or fflSTlHlE.
49. The use of claim 47, wherein the cancer is characterized by a genotype that satisfies Genetic Profile 1 of Figure 3.
50. The use of claim 47, wherein the cancer is characterized by a genotype that satisfies Genetic Profile 2 of Figure 3.
51. The use of claim 47, wherein the cancer comprises a MYD88 gain-of-function mutation.
52. The use of claim 51, wherein the cancer does not comprise an EZH2 gain-of-function mutation.
53. The use of any one of claims 47-52, wherein the at least one mutation decreases the function of a protein encoded by the mutated sequence as compared to the function of the protein encoded by the wild-type sequence.
54. The use of any one of claims 47-53, wherein the at least one mutation is a loss-of-function mutation.
55. The use of any one of claims 47-54, wherein the use further comprises detecting the at least one mutation in the subject.
56. The use of claim 55, wherein the detecting comprises subjecting a sample obtained from the subject to a sequence analysis assay.
57. The use of claim 56, wherein the analysis assay is a cobas® assay.
58. The use of claim 56 or 57, wherein the detecting comprises obtaining a sample from the subject.
59. The use of claim 58, wherein the sample is a tumor sample or a sample that comprises a cancer cell.
60. The use of claim 58, wherein the sample is a sample comprising ctDNA.
61. The use of any one of claims 47-60, wherein the inhibitor of EZH2 is (tazemetostat), or a pharmaceutically-acceptable salt thereof.
62. The use of any one of claims 47-61, wherein the inhibitor of EZH2 is administered orally.
63. The use of claim 62, wherein the inhibitor of EZH2 is formulated as a tablet.
64. The use of any one of claims 47-63, wherein the therapeutically effective amount of the inhibitor of EZH2 is between 100 mg and 3200 mg per day.
65. The use of claim 64, wherein the therapeutically effective amount of the inhibitor of EZH2 is 100 mg, 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, 1400 mg, 1600 mg or 3200 mg per day.
66. The use of claim 65, wherein the therapeutically effective amount is 1600 mg per day.
67. The use of any one of claims 47-66, wherein the cancer expresses a wild type EZH2 protein and does not express a mutant EZH2 protein.
68. The use of any one of claims 47-66, wherein the cancer expresses a mutant EZH2 protein.
69. The use of claim 68, wherein the mutant EZH2 protein comprises a substitution of any amino acid other than tyrosine (Y) for tyrosine (Y) at position 641 of SEQ ID NO: 1.
70. The use of claim 68 or 69, wherein the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 682 of SEQ ID NO: 1.
71. The use of any one of claims 68-70, wherein the mutant EZH2 protein comprises a substitution of any amino acid other than alanine (A) for alanine (A) at position 692 of SEQ ID NO: 1.
72. The use of any one of claims 47-71, wherein the at least one mutation comprises a MYD88 and/or STAT6A mutation, preferably wherein the MYD88 mutation is a gain-of-function mutation, preferably a MYD88 L273 mutation.
73. The use of any one of claims 47-72, wherein the cancer does not have a MYC and/or a HIST1H1E mutation.
74. The use of any one of claims 47-73, wherein the cancer (a) has a MYD88 gain-of-function or a STAT6A loss-of-function mutation, and (b) has neither a MYC nor a HIST1H1E mutation.
75. The use of any one of claims 47-74, wherein the cancer (a) has a MYD88 L273 mutation or a STAT6A loss-of-function mutation, (b) has neither a MYC nor a HIST1H1E mutation, and (c) does not have an EZH2 gain-of-function mutation.
76. The use of any one of claims 47-75, wherein the subject is a human subject.
77. The use of any one of claims 47-76, wherein the subject has cancer.
78. The use of claim 77, wherein the cancer is B-cell lymphoma.
79. The use of claim 78, wherein the B-cell lymphoma is an activated B-cell (ABC) type.
80. The use of claim 78, wherein the B-cell lymphoma is a germinal B-cell (GBC) type.
81. The use of claim 77, wherein the cancer is follicular lymphoma.
82. Use of an EZH2 inhibitor for treating a subject in need, the use comprising selecting a subject having cancer for treatment based on the presence of at least one mutation associated with a positive response to such treatment in the subject and/or based on the absence of at least one mutation associated with no response or with a negative response to such treatment in the subject.
83. The use of claim 82, wherein the at least one mutation associated with a positive response comprise
(a) an EZH2 gain-of-function mutation or a MYD88 gain-of-function mutation; and,
(b) a STAT6 loss-of-function mutation.
84. The use of any one of claims 82 or 83, wherein the at least one mutation associated with no response or with a negative response comprise
(a) a MYC mutation;
(b) a TP53 mutation; and/or,
(c) a HIST1H1E mutation.
85. The use of any one of claims 83-84, wherein the use comprises detecting the at least one mutation associated with a positive response and/or the at least one mutation associated with no response or a negative response in a sample obtained from the subject.
86. The use of any one of claims 83-85, wherein the use comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject
(a) having either an EZH2 or a MYD88 gain-of-function mutation, or a STAT6A loss-of-function mutation; and,
(b) having neither a MYC mutation, nor a TP53 mutation, nor a HIST1H1E mutation.
87. The use of any one of claims 83-86, wherein the use comprises selecting the subject for treatment with the EZH2 inhibitor based on the subject
(a) having a MYD88 L273 mutation or a STAT6A mutation; and,
(b) having neither a MYC mutation, nor a TP53 mutation, nor a HIST 1 HIE mutation.
88. The use of any one of claims 47-87, wherein the at least one mutation consists of a single mutation.
89. The use of any one of claims 47-87, wherein the at least one mutation comprises 2 mutations or more.
90. The use of any one of claims 47-89, wherein the at least one mutation comprises 3 mutations or more.
91. The use of any one of claims 47-90, wherein the at least one mutation comprises 4 mutations or more.
92. Use of an EZH2 inhibitor for treating a subject in need, the use comprising selecting a subject having cancer for treatment based on the presence of a mutation profile in the subject that matches a mutation profile of a patient exhibiting a complete or partial response or stable disease in any of Tables 1-5 or Figures 1-3.
PCT/US2018/037321 2017-06-13 2018-06-13 Inhibitors of ezh2 and methods of use thereof WO2018231973A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/622,391 US20230201212A1 (en) 2017-06-13 2018-06-13 Inhibitors of ezh2 and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762519078P 2017-06-13 2017-06-13
US62/519,078 2017-06-13

Publications (1)

Publication Number Publication Date
WO2018231973A1 true WO2018231973A1 (en) 2018-12-20

Family

ID=62817087

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/037321 WO2018231973A1 (en) 2017-06-13 2018-06-13 Inhibitors of ezh2 and methods of use thereof

Country Status (2)

Country Link
US (1) US20230201212A1 (en)
WO (1) WO2018231973A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
US11326212B2 (en) 2010-06-23 2022-05-10 British Columbia Cancer Agency Branch Biomarkers for non-hodgkin lymphomas and uses thereof
US11452727B2 (en) 2017-09-05 2022-09-27 Epizyme, Inc. Combination therapy for treating cancer

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
WO2011140324A1 (en) 2010-05-07 2011-11-10 Glaxosmithkline Llc Indoles
US8410088B2 (en) 2011-04-13 2013-04-02 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
WO2013120104A2 (en) 2012-02-10 2013-08-15 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
WO2014124418A1 (en) 2013-02-11 2014-08-14 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US20140378470A1 (en) * 2011-09-30 2014-12-25 GlaxoSmithKline L.L.C., a company Methods of treating cancer
EP2914296A1 (en) * 2012-11-01 2015-09-09 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
US20170065628A1 (en) * 2010-09-10 2017-03-09 Epizyme, Inc. Inhibitors of Human EZH2, and Methods of Use Thereof

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
WO2011140324A1 (en) 2010-05-07 2011-11-10 Glaxosmithkline Llc Indoles
US8536179B2 (en) 2010-05-07 2013-09-17 Glaxosmithkline Llc Indoles
US20170065628A1 (en) * 2010-09-10 2017-03-09 Epizyme, Inc. Inhibitors of Human EZH2, and Methods of Use Thereof
US8410088B2 (en) 2011-04-13 2013-04-02 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
US8765732B2 (en) 2011-04-13 2014-07-01 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
US9090562B2 (en) 2011-04-13 2015-07-28 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
US20140378470A1 (en) * 2011-09-30 2014-12-25 GlaxoSmithKline L.L.C., a company Methods of treating cancer
WO2013120104A2 (en) 2012-02-10 2013-08-15 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
US20150368229A1 (en) 2012-02-10 2015-12-24 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
EP2914296A1 (en) * 2012-11-01 2015-09-09 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
WO2014124418A1 (en) 2013-02-11 2014-08-14 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof

Non-Patent Citations (216)

* Cited by examiner, † Cited by third party
Title
"G enBank", Database accession no. NP_694543
"GenBank", Database accession no. AF232001. 1
"GenBank", Database accession no. NM _ 001982.3
"GenBank", Database accession no. NM _ 004656.3
"GenBank", Database accession no. NM __ 001007792.1
"GenBank", Database accession no. NM __ 004333.4
"GenBank", Database accession no. NM ___ 004064.4
"GenBank", Database accession no. NM ___ 004329.2
"GenBank", Database accession no. NM _000051.3
"GenBank", Database accession no. NM _000059.3
"GenBank", Database accession no. NM _000455.4
"GenBank", Database accession no. NM _000551.3
"GenBank", Database accession no. NM _000633.2
"GenBank", Database accession no. NM _001042594.1
"GenBank", Database accession no. NM _001122607.1
"GenBank", Database accession no. NM _001164619.1
"GenBank", Database accession no. NM _001172566.1
"GenBank", Database accession no. NM _001178133.1
"GenBank", Database accession no. NM _001243744.1
"GenBank", Database accession no. NM _001308824.1
"GenBank", Database accession no. NM _001321857.1
"GenBank", Database accession no. NM _001429.3
"GenBank", Database accession no. NM _002020.4
"GenBank", Database accession no. NM _002880.3
"GenBank", Database accession no. NM _003286.3
"GenBank", Database accession no. NM _004343.3
"GenBank", Database accession no. NM _004629.1
"GenBank", Database accession no. NM _005321.2
"GenBank", Database accession no. NM _005359.5
"GenBank", Database accession no. NM _006231.3
"GenBank", Database accession no. NM _023067.3
"GenBank", Database accession no. NM _181042.4
"GenBank", Database accession no. NM 000044.3
"GenBank", Database accession no. NM 000179.2
"GenBank", Database accession no. NM 000314.6
"GenBank", Database accession no. NM 000321.2
"GenBank", Database accession no. NM 000377.2
"GenBank", Database accession no. NM 000380.3
"GenBank", Database accession no. NM 000546.5
"GenBank", Database accession no. NM 000876.3
"GenBank", Database accession no. NM 001007468.2
"GenBank", Database accession no. NM 001014431.1
"GenBank", Database accession no. NM 001024847.2
"GenBank", Database accession no. NM 001042492.2
"GenBank", Database accession no. NM 001048171.1
"GenBank", Database accession no. NM 001079846
"GenBank", Database accession no. NM 001130442.2
"GenBank", Database accession no. NM 001130867.1
"GenBank", Database accession no. NM 001135937.2
"GenBank", Database accession no. NM 001145662.1
"GenBank", Database accession no. NM 001163213.1
"GenBank", Database accession no. NM 001173078.1
"GenBank", Database accession no. NM 001243027.2
"GenBank", Database accession no. NM 001278433.1
"GenBank", Database accession no. NM 001280552.1
"GenBank", Database accession no. NM 001281435.1
"GenBank", Database accession no. NM 001282387.1
"GenBank", Database accession no. NM 001288705.2
"GenBank", Database accession no. NM 001291241.1
"GenBank", Database accession no. NM 001293231.1
"GenBank", Database accession no. NM 001300734.1
"GenBank", Database accession no. NM 001308632.1
"GenBank", Database accession no. NM 001319984.1
"GenBank", Database accession no. NM 002468.4
"GenBank", Database accession no. NM 002769.4
"GenBank", Database accession no. NM 002944.2
"GenBank", Database accession no. NM 003153.4
"GenBank", Database accession no. NM 003924.3
"GenBank", Database accession no. NM 004260.3
"GenBank", Database accession no. NM 004628.4
"GenBank", Database accession no. NM 005120.2
"GenBank", Database accession no. NM 005157
"GenBank", Database accession no. NM 005417.4
"GenBank", Database accession no. NM 005631.4
"GenBank", Database accession no. NM 014757.4
"GenBank", Database accession no. NM 015338.5
"GenBank", Database accession no. NM 017628.4
"GenBank", Database accession no. NM 017841.2
"GenBank", Database accession no. NM 022725.3
"GenBank", Database accession no. NM 024529.4
"GenBank", Database accession no. NM 033360.3
"GenBank", Database accession no. NM 078626.2
"GenBank", Database accession no. NM 080425.3
"GenBank", Database accession no. NM_ 001289936.1
"GenBank", Database accession no. NM_ 001320893.1
"GenBank", Database accession no. NM_ 006206.4
"GenBank", Database accession no. NM_ 017617.4
"GenBank", Database accession no. NM_ 021922.2
"GenBank", Database accession no. NM_000038.5
"GenBank", Database accession no. NM_000057.3
"GenBank", Database accession no. NM_000075.3
"GenBank", Database accession no. NM_000123.3
"GenBank", Database accession no. NM_000127.2
"GenBank", Database accession no. NM_000141.4
"GenBank", Database accession no. NM_000143.3
"GenBank", Database accession no. NM_000215.3
"GenBank", Database accession no. NM_000245.3
"GenBank", Database accession no. NM_000249.3
"GenBank", Database accession no. NM_000251.2
"GenBank", Database accession no. NM_000264.3
"GenBank", Database accession no. NM_000369.2
"GenBank", Database accession no. NM_000378.4
"GenBank", Database accession no. NM_000435.2
"GenBank", Database accession no. NM_000489.4
"GenBank", Database accession no. NM_000535.6
"GenBank", Database accession no. NM_000545.6
"GenBank", Database accession no. NM_000553.4
"GenBank", Database accession no. NM_001 166049.1
"GenBank", Database accession no. NM_001014796.1
"GenBank", Database accession no. NM_001024688.2
"GenBank", Database accession no. NM_001033081.2
"GenBank", Database accession no. NM_001042355.1
"GenBank", Database accession no. NM_001079668.2
"GenBank", Database accession no. NM_001079846.1
"GenBank", Database accession no. NM_001093772. 1
"GenBank", Database accession no. NM_001098209.1
"GenBank", Database accession no. NM_001105.4
"GenBank", Database accession no. NM_001145104.1
"GenBank", Database accession no. NM_001145306.1
"GenBank", Database accession no. NM_001145337.2
"GenBank", Database accession no. NM_001162427.1
"GenBank", Database accession no. NM_001163147.1
"GenBank", Database accession no. NM_001163287.1
"GenBank", Database accession no. NM_001172567.1
"GenBank", Database accession no. NM_001172568.1
"GenBank", Database accession no. NM_001172569.1
"GenBank", Database accession no. NM_001174087.1
"GenBank", Database accession no. NM_001178079.1
"GenBank", Database accession no. NM_001178080.1
"GenBank", Database accession no. NM_001178081.1
"GenBank", Database accession no. NM_001193376.1
"GenBank", Database accession no. NM_001195132.1
"GenBank", Database accession no. NM_001200001.1
"GenBank", Database accession no. NM_001211.5
"GenBank", Database accession no. NM_001270508.1
"GenBank", Database accession no. NM_001276503.1
"GenBank", Database accession no. NM_001278519.1
"GenBank", Database accession no. NM_001282418.1
"GenBank", Database accession no. NM_001285829.1
"GenBank", Database accession no. NM_001286167.1
"GenBank", Database accession no. NM_001287344.1
"GenBank", Database accession no. NM_001290114.1
"GenBank", Database accession no. NM_001291628.1
"GenBank", Database accession no. NM_001291847.1
"GenBank", Database accession no. NM_001291858.1
"GenBank", Database accession no. NM_001291970.1
"GenBank", Database accession no. NM_001303418.1
"GenBank", Database accession no. NM_001308133.1
"GenBank", Database accession no. NM_001318832.1
"GenBank", Database accession no. NM_001321051.1
"GenBank", Database accession no. NM_001322195.1
"GenBank", Database accession no. NM_001322262.1
"GenBank", Database accession no. NM_001330437.1
"GenBank", Database accession no. NM_001346900.1
"GenBank", Database accession no. NM_001429.3
"GenBank", Database accession no. NM_002049.3
"GenBank", Database accession no. NM_002067.4
"GenBank", Database accession no. NM_002072.4
"GenBank", Database accession no. NM_002107.4
"GenBank", Database accession no. NM_002253.2
"GenBank", Database accession no. NM_002467.4
"GenBank", Database accession no. NM_002520.6
"GenBank", Database accession no. NM_002755.3
"GenBank", Database accession no. NM_003600.3
"GenBank", Database accession no. NM_003745.1
"GenBank", Database accession no. NM_004119.2
"GenBank", Database accession no. NM_004304.4
"GenBank", Database accession no. NM_004380
"GenBank", Database accession no. NM_004456
"GenBank", Database accession no. NM_004655.3
"GenBank", Database accession no. NM_005188.3
"GenBank", Database accession no. NM_005235.2
"GenBank", Database accession no. NM_005324.4
"GenBank", Database accession no. NM_005373.2
"GenBank", Database accession no. NM_006015.4
"GenBank", Database accession no. NM_006218.3
"GenBank", Database accession no. NM_006297.2
"GenBank", Database accession no. NM_007294.3
"GenBank", Database accession no. NM_015125.4
"GenBank", Database accession no. NM_016038.2
"GenBank", Database accession no. NM_017763.5
"GenBank", Database accession no. NM_018193.2
"GenBank", Database accession no. NM_018315.4
"GenBank", Database accession no. NM_020732.3
"GenBank", Database accession no. NM_024675.3
"GenBank", Database accession no. NM_032043.21
"GenBank", Database accession no. NM_053056.2
"GenBank", Database accession no. NM_078487.2
"GenBank", Database accession no. NM_130804.2
"GenBank", Database accession no. NM_144606.5
"GenBank", Database accession no. NM_145862.2
"GenBank", Database accession no. NM_152998
"GenBank", Database accession no. NM_181831.2
"GenBank", Database accession no. NM_213647.2
"GenBank", Database accession no. NP _001073315.1
"GenBank", Database accession no. NP 004447
"GenBank", Database accession no. NP_001420.2
"GenBank", Database accession no. NP_004371
"GenBank", Database accession no. NP_004447
"GenBank", Database accession no. NP_694543
"GenBank", Database accession no. NR_103873.1
"GenBank", Database accession no. X02596.1
"Remington: the Science and Practice of Pharmacy", 1995, MACK PUBLISHING CO.
BUNDEGAARD, H.: "Design of Prodrugs", 1985, ELESEVIER, pages: 92
CAHN ET AL., ANGEW. CHEM. INTER. EDIT., vol. 5, 1966, pages 385
CAHN ET AL., ANGEW. CHEM., vol. 78, 1966, pages 413
CAHN ET AL., EXPERIENTIA, vol. 12, 1956, pages 81
CAHN, J CHEM. EDUC., vol. 41, 1964, pages 116
CAHN; INGOLD, J. CHEM. SOC., vol. 612, 1951
CHEN ET AL., GENOMICS, vol. 38, 1996, pages 30 - 7
KNUTSON, NAT. CHEM. BIOL., vol. 8, 2012, pages 890 - 896
M. TESTONI ET AL: "Genetic lesions in diffuse large B-cell lymphomas", ANNALS OF ONCOLOGY., vol. 26, no. 6, 20 January 2015 (2015-01-20), NL, pages 1069 - 1080, XP055361604, ISSN: 0923-7534, DOI: 10.1093/annonc/mdv019 *
MORIN ET AL., NAT GENET, vol. 42, no. 2, 2010, pages 181 - 5
PATANI; LAVOIE, CHEM. REV., vol. 96, 1996, pages 3147 - 3176
S. K. KNUTSON ET AL: "Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma", MOLECULAR CANCER THERAPEUTICS, vol. 13, no. 4, 1 April 2014 (2014-04-01), US, pages 842 - 854, XP055247668, ISSN: 1535-7163, DOI: 10.1158/1535-7163.MCT-13-0773 *
V RIBRAG: "Phase 1 Study of Tazemetostat (EPZ-6438), an Inhibitor of Enhancer of Zeste-Homolog 2 (EZH2): Preliminary Safety and Activity in Relapsed or Refractory Non-Hodgkin Lymphoma (NHL) Patients", 57TH ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY; ORLANDO, FL, USA, 3 December 2015 (2015-12-03), XP055362197 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11326212B2 (en) 2010-06-23 2022-05-10 British Columbia Cancer Agency Branch Biomarkers for non-hodgkin lymphomas and uses thereof
US11452727B2 (en) 2017-09-05 2022-09-27 Epizyme, Inc. Combination therapy for treating cancer
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods

Also Published As

Publication number Publication date
US20230201212A1 (en) 2023-06-29

Similar Documents

Publication Publication Date Title
US20220193084A1 (en) Inhibitors of ezh2 and methods of use thereof
US11642347B2 (en) EZH2 inhibitors for treating lymphoma
AU2016325643B2 (en) Method of treating malignant rhabdoid tumor or small cell cancer of the ovary of the hypercalcemic type
WO2018231973A1 (en) Inhibitors of ezh2 and methods of use thereof
US20180235975A1 (en) Method for treating cancer
JP2018536009A5 (en)
US11738018B2 (en) Inhibiting mutant isocitrate dehydrogenase 1 (mIDH-1)
NZ727108B2 (en) Ezh2 inhibitors for treating lymphoma

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18737471

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18737471

Country of ref document: EP

Kind code of ref document: A1